US9388465B2 - Polynucleotide barcode generation - Google Patents

Polynucleotide barcode generation Download PDF

Info

Publication number
US9388465B2
US9388465B2 US14/175,973 US201414175973A US9388465B2 US 9388465 B2 US9388465 B2 US 9388465B2 US 201414175973 A US201414175973 A US 201414175973A US 9388465 B2 US9388465 B2 US 9388465B2
Authority
US
United States
Prior art keywords
polynucleotides
polynucleotide
partitions
capsule
partition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US14/175,973
Other versions
US20140228255A1 (en
Inventor
Benjamin Hindson
Mirna Jarosz
Paul Hardenbol
Michael Schnall-Levin
Kevin Ness
Serge Saxonov
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
10X Genomics Inc
Original Assignee
10X Genomics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51297676&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US9388465(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US14/175,973 priority Critical patent/US9388465B2/en
Application filed by 10X Genomics Inc filed Critical 10X Genomics Inc
Priority to US14/250,701 priority patent/US20140235506A1/en
Assigned to 10X TECHNOLOGIES, INC. reassignment 10X TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JAROSZ, MIRNA, HARDENBOL, PAUL, HINDSON, BENJAMIN, NESS, KEVIN, SAXONOV, SERGE, SCHNALL-LEVIN, MICHAEL
Publication of US20140228255A1 publication Critical patent/US20140228255A1/en
Assigned to 10X GENOMICS, INC. reassignment 10X GENOMICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: 10X TECHNOLOGIES, INC.
Priority to US15/200,928 priority patent/US20160304860A1/en
Publication of US9388465B2 publication Critical patent/US9388465B2/en
Application granted granted Critical
Priority to US15/376,582 priority patent/US9701998B2/en
Priority to US15/588,519 priority patent/US9856530B2/en
Priority to US15/850,241 priority patent/US10676789B2/en
Priority to US15/980,473 priority patent/US10253364B2/en
Priority to US16/000,803 priority patent/US20180282804A1/en
Priority to US16/052,431 priority patent/US10273541B2/en
Priority to US16/052,486 priority patent/US10323279B2/en
Priority to US16/165,389 priority patent/US10533221B2/en
Priority to US16/212,441 priority patent/US10752949B2/en
Priority to US16/231,142 priority patent/US10584381B2/en
Priority to US16/231,185 priority patent/US10400280B2/en
Priority to US16/246,322 priority patent/US10597718B2/en
Priority to US16/294,769 priority patent/US10450607B2/en
Priority to US16/395,090 priority patent/US10669583B2/en
Priority to US16/435,362 priority patent/US10626458B2/en
Priority to US16/435,417 priority patent/US10752950B2/en
Priority to US16/698,740 priority patent/US11473138B2/en
Priority to US16/736,323 priority patent/US20200255894A1/en
Priority to US16/844,141 priority patent/US11441179B2/en
Priority to US16/852,906 priority patent/US11421274B2/en
Priority to US16/998,425 priority patent/US11035002B2/en
Priority to US16/998,414 priority patent/US11021749B2/en
Priority to US17/314,526 priority patent/US11359239B2/en
Priority to US17/392,610 priority patent/US20220098659A1/en
Priority to US18/186,088 priority patent/US20240002929A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1065Preparation or screening of tagged libraries, e.g. tagged microorganisms by STM-mutagenesis, tagged polynucleotides, gene tags
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/5436Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals with ligand physically entrapped within the solid phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2563/00Nucleic acid detection characterized by the use of physical, structural and functional properties
    • C12Q2563/179Nucleic acid detection characterized by the use of physical, structural and functional properties the label being a nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification

Definitions

  • Polynucleotide barcodes have utility in numerous applications, including next generation sequencing techniques. Such barcodes generally contain unique identifier sequences, which can be extremely expensive to manufacture at sufficient diversity and scale.
  • the cost of synthesizing a single polynucleotide barcode is a function of the cost per base during synthesis and the length of the polynucleotide.
  • the cost of synthesizing a plurality of barcodes, each with a different sequence is therefore equivalent to the cost per base, multiplied by the number of bases per molecule, multiplied by the number of molecules within the plurality of barcodes.
  • this cost is prohibitive.
  • This disclosure provides methods, compositions, systems, and kits for the generation of polynucleotide barcodes and the use of such polynucleotide barcodes.
  • polynucleotide barcodes may be used for any suitable application.
  • An aspect of the disclosure provides a library comprising one or more polynucleotides, each of the polynucleotides comprising a barcode sequence, wherein the polynucleotides are disposed within one or more partitions, and wherein the library comprises at least about 1,000 different barcode sequences.
  • the barcode sequences are at least about 5 nucleotides in length. Also, the barcode sequences may be random polynucleotide sequences.
  • the partitions may comprise, on average, about 1 polynucleotide, about 0.5 polynucleotides, or about 0.1 polynucleotides.
  • the partitions may be droplets, capsules, wells or beads.
  • the library may comprise at least about 10,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 500,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 2,500,00 different barcode sequences, at least about 5,000,000 different barcode sequences, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
  • the partitions may comprise multiple copies of the same polynucleotide.
  • each of the polynucleotides may comprise a sequence selected from the group consisting of an immobilization sequence, an annealing sequence for a sequencing primer, and a sequence compatible for ligation with a target polynucleotide.
  • each of the polynucleotides is a MALBAC primer.
  • Another aspect of the disclosure provides a method of synthesizing a library of polynucleotides comprising barcode sequences, the method comprising: a.) synthesizing a plurality of polynucleotides comprising barcode sequences; b.) separating the polynucleotides into a plurality of partitions, thereby generating partitioned polynucleotides; c.) amplifying the partitioned polynucleotides, thereby generating amplified polynucleotides; and d.) isolating partitions comprising amplified polynucleotides.
  • the synthesizing comprises including a mixture of adenine, thymine, guanine, and cytosine in a coupling reaction.
  • the separating may comprise performing a limiting dilution, thereby generating diluted polynucleotides. In some cases, the separating further comprises partitioning said diluted polynucleotides.
  • the amplifying may be performed by a method selected from the group consisting of polymerase chain reaction, asymmetric polymerase chain reaction, emulsion PCR (ePCR), ePCR including the use of a bead, ePCR including the use of a hydrogel, multiple annealing and looping-based amplification cycles (MALBAC), single primer isothermal amplification, and combinations thereof.
  • ePCR emulsion PCR
  • MALBAC multiple annealing and looping-based amplification cycles
  • the amplifying is performed using an RNA primer and may include exposing the amplified polynucleotides to an RNAase H.
  • each of said polynucleotides comprising barcode sequences is a MALBAC primer.
  • the isolating may be performed by flow-assisted sorting.
  • a hairpin structure may be formed from a polynucleotide selected from the group consisting of the polynucleotides comprising barcode sequences and the amplified polynucleotides.
  • a method may further comprise cutting the hairpin structure within an unannealed region.
  • a polynucleotide selected from the group consisting of said polynucleotides comprising barcode sequences, said partitioned polynucleotides, and said amplified polynucleotides may be attached to a bead.
  • the method may further comprise annealing the amplified polynucleotides with a partially complementary sequence.
  • the partially complementary sequence may comprise a barcode sequence.
  • the method may further comprise attaching at least one of the amplified polynucleotides to a target sequence.
  • the target sequence may be fragmented.
  • the target sequence is fragmented by a method selected from the group consisting of mechanical shear and treatment with an enzyme.
  • the mechanical shear may be induced by ultrasound.
  • the enzyme is selected from the group consisting of a restriction enzyme, a fragmentase, and a transposase.
  • the attaching may be performed by a method selected from the group consisting of ligation and amplification.
  • the amplification is a MALBAC amplification performed with MALBAC primers, thereby generating a MALBAC amplification product.
  • the MALBAC primers comprise the amplified polynucleotides.
  • the MALBAC primers comprise polynucleotides that are not said amplified polynucleotides.
  • the method may further comprise attaching the MALBAC amplification product to the amplified polynucleotide.
  • each of the partitions may comprise, on average, about 1 polynucleotide comprising a barcode sequence, 0.5 polynucleotides comprising barcode sequences, or 0.1 polynucleotides comprising barcode sequences.
  • the partitions may be selected from the group consisting of droplets, capsules, and wells.
  • the library comprises at least about 1,000 different barcode sequences, at least about 10,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 500,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 2,500,00 different barcode sequences, at least about 5,000,000 different barcode sequences, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
  • the partitions comprise multiple copies of the same polynucleotide comprising a barcode sequence.
  • the polynucleotides comprising barcode sequences may comprise a sequence selected from the group consisting of an immobilization sequence, an annealing sequence for a sequencing primer, and a sequence compatible for ligation with a target polynucleotide.
  • An additional aspect of the disclosure provides a library comprising at least about 1,000 beads, wherein each bead of the at least about 1,000 beads comprises a different barcode sequence.
  • the different barcode sequence can be included in a polynucleotide comprising an immobilization sequence and/or an annealing sequence for a sequencing primer.
  • the different barcode sequence can be at least about 5 nucleotides or at least about 10 nucleotides in length.
  • the different barcode sequence can be a random polynucleotide sequence or can be generated combinatorially.
  • each of the 1,000 beads can comprise multiple copies of the different barcode sequence.
  • each of the 1,000 beads may comprise at least about 100,000, at least about 1,000,000, or at least about 10,000,000 copies of the different barcode sequence.
  • the library can further comprise two or more beads comprising the same barcode sequence.
  • at least two beads of the 1,000 beads can comprise the same barcode sequence.
  • the at least about 1,000 beads may comprise at least about 10,000 beads, or at least about 100,000 beads.
  • the library can comprise at least about 1,000, at least about 10,000, at least about 100,000, at least about 1,000,000, at least about 2,500,000, at least about 5,000,000, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
  • the at least about 1,000 beads can be distributed across a plurality of partitions.
  • the partitions can be droplets of an emulsion.
  • each bead of the 1,000 beads can be included in a different partition.
  • the different partition can be a droplet of an emulsion.
  • two or more beads of the 1,000 beads can be included in a different partition.
  • the different partition can be a droplet of an emulsion.
  • the 1,000 beads can be hydrogel beads.
  • An additional aspect of the disclosure provides for use of a library, composition, method, device, or kit described herein in partitioning species, in partitioning oligonucleotides, in stimulus-selective release of species from partitions, in performing reactions (e.g., ligation and amplification reactions) in partitions, in performing nucleic acid synthesis reactions, in barcoding nucleic acid, in preparing polynucleotides for sequencing, in sequencing polynucleotides, in mutation detection, in neurologic disorder diagnostics, in diabetes diagnostics, in fetal aneuploidy diagnostics, in cancer mutation detection and forenscics, in disease detection, in medical diagnostics, in low input nucleic acid applications, in circulating tumor cell (CTC) sequencing, in polynucleotide phasing, in sequencing polynucleotides from small numbers of cells, in analyzing gene expression, in partitioning polynucleotides from cells, or in a combination thereof.
  • CTC circulating tumor cell
  • FIG. 1 is schematically depicts an example forked adapter.
  • FIG. 2 schematically depicts example placements of barcode regions.
  • FIG. 3 depicts example sequences of two forked adapters ligated to opposite ends of a target polynucleotide. Full-length sequence disclosed as SEQ ID NO: 35.
  • FIG. 4 is a schematic example method used to generate a forked adapter described in Example 1.
  • FIG. 5 is a schematic example of a capsule within a capsule described in Example 2.
  • FIG. 6 is a schematic example of capsules within a capsule described in Example 3.
  • FIG. 7 is a schematic example of a product (or intermediate) that may be generated according to methods of Example 4.
  • FIGS. 8 a - c depict example sequences described in Example 4.
  • FIGS. 9 a - j depict example sequences described in Example 5.
  • FIGS. 10 a - e depict example sequences described in Example 6.
  • FIGS. 11 a - d schematically depict methods and structures described in Example 7.
  • FIG. 12 schematically depicts the production capsules via an example flow-focusing method.
  • FIG. 13 schematically depicts the production of capsules within capsules via an example flow-focusing method.
  • FIGS. 14 a - e schematically depict methods and structures described in Example 8.
  • FIGS. 15 a - e schematically depict methods and structures described in Example 9.
  • FIG. 16 schematically depicts methods and structures described in Example 10.
  • FIG. 17 schematically depicts a capsule within a capsule described in Example 11.
  • FIG. 18 schematically depicts capsules within a capsule described in Example 12.
  • FIGS. 19 a - e depict example sequences described in Example 13.
  • FIG. 19 f describes example methods and structures described in Example 13.
  • FIG. 20 schematically depicts a capsule within a capsule described in Example 14.
  • FIGS. 21 a - c schematically depict methods and structures described in Example 15.
  • FIG. 22 schematically depicts a capsule within a capsule described in Example 16.
  • FIG. 23 schematically depicts a capsule within a capsule described in Example 17.
  • polynucleotide barcodes may be used for any suitable application.
  • the polynucleotide barcodes provided in this disclosure may be used in next generation sequencing reactions.
  • Next generation sequencing reactions include the sequencing of whole genomes, detection of specific sequences such as single nucleotide polymorphisms (SNPs) and other mutations, detection of nucleic acid (e.g., deoxyribonucleic acid) insertions, and detection of nucleic acid deletions.
  • SNPs single nucleotide polymorphisms
  • nucleic acid e.g., deoxyribonucleic acid
  • Utilization of the methods, compositions, systems, and kits described herein may incorporate, unless otherwise indicated, any conventional techniques of organic chemistry, polymer technology, microfluidics, molecular biology, recombinant techniques, cell biology, biochemistry, and immunology.
  • Such conventional techniques include well and microwell construction, capsule generation, generation of emulsions, spotting, microfluidic device construction, polymer chemistry, restriction digestion, ligation, cloning, polynucleotide sequencing, and polynucleotide sequence assembly.
  • suitable techniques are described throughout this disclosure. However, equivalent procedures may also be utilized. Descriptions of certain techniques may be found in standard laboratory manuals, such as Genome Analysis: A Laboratory Manual Series ( Vols.
  • barcode generally refers to a label that may be attached to an analyte to convey information about the analyte.
  • a barcode may be a polynucleotide sequence attached to fragments of a target polynucleotide contained within a particular partition. This barcode may then be sequenced with the fragments of the target polynucleotide. The presence of the same barcode on multiple sequences may provide information about the origin of the sequence. For example, a barcode may indicate that the sequence came from a particular partition and/or a proximal region of a genome. This may be particularly useful for sequence assembly when several partitions are pooled before sequencing.
  • base pairs generally refers to an abbreviation for “base pairs”.
  • microwell generally refers to a well with a volume of less than 1 mL. Microwells may be made in various volumes, depending on the application. For example, microwells may be made in a size appropriate to accommodate any of the partition volumes described herein.
  • partition may be a verb or a noun.
  • the term When used as a verb (e.g., “to partition,” or “partitioning”), the term generally refers to the fractionation (e.g., subdivision) of a species or sample (e.g., a polynucleotide) between vessels that can be used to sequester one fraction (or subdivision) from another. Such vessels are referred to using the noun “partition.” Partitioning may be performed, for example, using microfluidics, dilution, dispensing, and the like.
  • a partition may be, for example, a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a bead, a surface of a bead in dilute solution, or any other suitable container for sequestering one fraction of a sample from another.
  • a partition may also comprise another partition.
  • polynucleotide or “nucleic acid,” as used herein, generally refer to molecules comprising a plurality of nucleotides.
  • exemplary polynucleotides include deoxyribonucleic acids, ribonucleic acids, and synthetic analogues thereof, including peptide nucleic acids.
  • species generally refers to any substance that can be used with the methods, compositions, systems, devices, and kits of this disclosure.
  • species include reagents, analytes, cells, chromosomes, tagging molecules or groups of molecules, barcodes, and any sample comprising any of these species. Any suitable species may be used, as more fully discussed elsewhere in this disclosure.
  • Barcodes may be of a variety of different formats, including polynucleotide barcodes. Depending upon the specific application, barcodes may be attached to polynucleotide fragments in a reversible or irreversible manner. Additionally, barcodes may allow for identification and/or quantification of individual polynucleotide fragments during sequencing.
  • Barcodes may be loaded into partitions so that one or more barcodes are introduced into a particular partition.
  • each partition may contain a different set of barcodes. This may be accomplished by directly dispensing the barcodes into the partitions, or by placing the barcodes within a partition that is contained within another partition.
  • the barcodes may be loaded into the partitions at an expected or predicted ratio of barcodes per species to be barcoded (e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.). In some cases, the barcodes are loaded into partitions such that about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the barcodes are loaded into partitions such that more than about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species.
  • an expected or predicted ratio of barcodes per species to be barcoded e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.
  • the barcodes are loaded into partitions such that about 0.0001, 0.001, 0.1,
  • the barcodes are loaded in the partitions so that less than about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species.
  • barcodes may be copies of the same barcode, or may be different barcodes.
  • the attachment process may be designed to attach multiple identical barcodes to a single polynucleotide fragment, or multiple different barcodes to the polynucleotide fragment.
  • the methods provided herein may comprise loading a partition with the reagents necessary for the attachment of barcodes to polynucleotide fragments.
  • reagents including restriction enzymes, ligase enzymes, buffers, adapters, barcodes and the like may be loaded into a partition.
  • reagents including primers, DNA polymerases, dNTPs, buffers, barcodes and the like may be loaded into a partition.
  • transposon-mediated barcoding e.g., NEXTERA
  • reagents including a transposome i.e., transposase and transposon end complex
  • buffers i.e., buffers, and the like
  • reagents including a MALBAC primer, buffers, and the like may be loaded into a partition. As described throughout this disclosure, these reagents may be loaded directly into the partition, or via another partition.
  • Barcodes may be ligated to a polynucleotide fragment using sticky or blunt ends. Barcoded polynucleotide fragments may also be generated by amplifying a polynucleotide fragment with primers comprising barcodes. In some cases, MALBAC amplification of the polynucleotide fragment may be used to generate a barcoded polynucleotide fragment. A primer used for MALBAC may or may not comprise a barcode. In cases where a MALBAC primer does not comprise a barcode, the barcode may be added to MALBAC amplification products by other amplification methods, such as, for example, PCR. Barcoded polynucleotide fragments may also be generated using transposon-mediated methods. As with any other species discussed in this disclosure, these modules may be contained within the same or different partitions, depending on the needs of the assay or process.
  • barcodes may be assembled combinatorially, from smaller components designed to assemble in a modular format.
  • three modules, 1A, 1B, and 1C may be combinatorially assembled to produce barcode 1ABC.
  • Such combinatorial assembly may significantly reduce the cost of synthesizing a plurality of barcodes.
  • barcodes may be combinatorially assembled by mixing two oligonucleotides and hybridizing them to produce annealed or partially annealed oligonucleotides (e.g., forked adapters). These barcodes may comprise an overhang of one or more nucleotides, in order to facilitate ligation with polynucleotide fragments that are to be barcoded. In some cases, the 5′ end of the antisense strand may be phosphorylated in order to ensure double-stranded ligation.
  • modules may be assembled by, for example, mixing oligonucleotides A and B, A and C, A and D, B and C, B, and D, and so on.
  • the annealed oligonucleotides may also be synthesized as a single molecule with a hairpin loop that may be cut after ligation to the polynucleotide to be barcoded.
  • attachment of polynucleotides to each other may rely on hybridization-compatible overhangs.
  • the hybridization between A and T is often used to ensure ligation compatibility between fragments.
  • an A overhang may be created by treatment with an enzyme, such as a Taq polymerase.
  • a restriction enzyme may be used to create a cleavage product with a single base 3′ overhang which may be, for example, A or T.
  • restriction enzymes that leave a single base 3′ overhang include MnII, HphI, Hpy188I, HpyAV, HpyCH4III, MboII, BciVI, BmrI, AhdI, and XcmI.
  • different overhangs e.g., 5′ overhangs, overhangs of greater than a single base
  • Additional restriction enzymes that may be used to generate overhangs include BfuC1, Taq ⁇ I, BbVI, Bcc1, BceA1, BcoDI, BsmAI, and BsmFI.
  • this disclosure provides methods for the generation of partitioned barcode libraries and libraries produced according to such methods.
  • the methods provided herein combine random synthesis of DNA sequences, separation into partitions, amplification of separated sequences, and isolation of amplified separated sequences to provide a library of barcodes contained within partitions.
  • the methods described herein utilize random methods of polynucleotide synthesis, including random methods of DNA synthesis.
  • any combination of A, C, G, and/or T may be added to a coupling step so that each type of base in the coupling step is coupled to a subset of the product. If A, C, G, and T are present at equivalent concentrations, approximately one-quarter of the product will incorporate each base. Successive coupling steps, and the random nature of the coupling reaction, enable the generation of 4 n possible sequences, where n is the number of bases in the polynucleotide.
  • any suitable synthetic bases may also be used with the invention.
  • the bases included in each coupling step may be altered in order to synthesize a preferred product.
  • the number of bases present in each coupling step may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
  • the number of bases present in each coupling step may be at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
  • the number of bases present in each coupling step may be less than 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • the concentration of the individual bases may also be altered in order to synthesize the preferred product.
  • any base may be present at a concentration of about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base.
  • any base may be present at a concentration of at least about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base.
  • any base may be present at a concentration of less than about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base.
  • the length of the random polynucleotide sequence may be any suitable length, depending on the application. In some cases, the length of the random polynucleotide sequence may be 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides. In some cases, the length of the random polynucleotide sequence may be at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides. In some cases, the length of the random polynucleotide sequence may be less than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides.
  • the library is defined by the number of members.
  • a library may comprise about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*10 11 , or 1.09951*10 12 members.
  • a library may comprise at least about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*10, or 1.09951*10 12 members.
  • a library may comprise less than about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*10, or 1.09951*10 12 members.
  • the library is a barcode library.
  • a barcode library may comprise at least about 1000, 10000, 100000, 1000000, 2500000, 5000000, 10000000, 25000000, 50000000, or 100000000 different barcode sequences.
  • the random barcode libraries may also comprise other polynucleotide sequences.
  • these other polynucleotide sequences are non-random in nature and include, for example, primer binding sites, annealing sites for the generation of forked adapters, immobilization sequences, and regions that enable annealing with a target polynucleotide sequence, and thus barcoding of the polynucleotide sequence.
  • the polynucleotides are partitioned into separate compartments to generate a library of partitioned polynucleotides comprising barcode sequences. Any suitable method of separation and any suitable partition or partitions within partitions may be used.
  • partitioning is performed by diluting the mixture of polynucleotides comprising random barcode sequences such that a particular volume of the dilution contains, on average, less than a single polynucleotide. The particular volume of the dilution may then be transferred to a partition. In any plurality of partitions, each partition is therefore likely to have one or zero polynucleotide molecules.
  • a dilution may be performed such that each partition comprises about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, or more molecules. In some cases a dilution may be performed such that each partition comprises at least about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, or more molecules. In some cases a dilution may be performed such that each partition comprises less than about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, or 2 molecules.
  • about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules. In some cases, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules. In some cases, less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules.
  • about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides. In some cases, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides. In some cases, less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides.
  • a partition is a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a surface of a bead, or any other suitable container for sequestering one fraction of a sample from another.
  • a partition includes a bead, a primer for amplification may be attached to the bead. Partitions are described in greater detail elsewhere in this disclosure.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • LATE-PCR linear after the exponential PCR
  • DOP-PCR degenerate oligonucleotide primer PCR
  • PEP-PCR primer extension pre-amplification PCR
  • MDA multiple displacement amplification
  • SPIA single primer isothermal amplification
  • emulsion PCR ePCR
  • ePCR emulsion PCR
  • MALBAC multiple emulsion PCR
  • MALBAC multiple annealing and looping-based amplification cycles
  • amplification methods that generate single-stranded product may be preferred, for example.
  • amplification methods that generate double-stranded products e.g., standard PCR
  • an amplification method will exponentially amplify the partitioned polynucleotide.
  • an amplification method will linearly amplify the partitioned polynucleotide.
  • an amplification method will first exponentially and then linearly amplify a polynucleotide.
  • a single type of amplification may be used to amplify polynucleotides or amplification may be completed with sequential steps of different types of amplification.
  • ePCR may be combined with further rounds of ePCR or may be combined with a different type of amplification.
  • Amplification is performed until a suitable amount of polynucleotide comprising a barcode is produced. In some cases, amplification may be performed for 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, or more cycles. In some cases, amplification may be performed for at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, or more cycles. In some cases, amplification may be performed for less than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 cycles.
  • amplification may be performed until a certain amount of polynucleotide product is produced in each partition. In some cases, amplification is performed until the amount of polynucleotide product is about 10,000,000,000; 5,000,000,000; 1,000,000,000; 500,000,000; 100,000,000; 50,000,000; 10,000,000; 5,000,000; 1,000,000; 500,000; 400,000; 300,000; 200,000; or 100,000 molecules. In some cases, amplification is performed until the amount of polynucleotide product is at least about 100,000; 200,000; 300,000; 400,000; 500,000; 1,000,000; 5,000,000; 10,000,000; 50,000,000; 100,000,000; 500,000,000; 1,000,000,000; 5,000,000,000; or 10,000,000,000 molecules.
  • amplification is performed until the amount of polynucleotide product is less than about 10,000,000,000; 5,000,000,000; 1,000,000,000; 500,000,000; 100,000,000; 50,000,000; 10,000,000; 5,000,000; 1,000,000; 500,000; 400,000; 300,000; 200,000; or 100,000 molecules.
  • polynucleotides comprising barcodes are partitioned such that each partition contains, on average, less than one polynucleotide sequence. Therefore, in some cases, a fraction of the partitions will not contain a polynucleotide and therefore cannot contain an amplified polynucleotide. Thus, it may be desirable to separate partitions comprising polynucleotides from partitions not comprising polynucleotides.
  • partitions comprising polynucleotides are separated from partitions not comprising polynucleotides using flow-based sorting methods capable of identifying partitions comprising polynucleotides.
  • an indicator of the presence of a polynucleotide may be used in order to differentiate partitions comprising polynucleotides from those not comprising polynucleotides.
  • a nucleic acid stain may be used to identify partition comprising polynucleotides.
  • exemplary stains include intercalating dyes, minor-groove binders, major groove binders, external binders, and bis-intercalators.
  • dyes include SYBR green, SYBR blue, DAPI, propidium iodide, SYBR gold, ethidium bromide, acridines, proflavine, acridine orange, acriflavine, fluorcoumanin, ellipticine, daunomycin, chloroquine, distamycin D, chromomycin, homidium, mithramycin, ruthenium polypyridyls, anthramycin, phenanthridines and acridines, ethidium bromide, propidium iodide, hexidium iodide, dihydroethidium, ethidium homodimer-1 and -2, ethidium monoazide, ACMA, indoles, imidazoles (e.g., Hoechst 33258, Hoechst 33342, Hoechst 34580 and DAPI), acridine orange (also capable
  • isolation methods such as magnetic separation or sedimentation of particles may be used. Such methods may include, for example, a step of attaching a polynucleotide to be amplified, a primer corresponding to said polynucleotide to be amplified, and/or a polynucleotide product of amplification to a bead.
  • attachment of a polynucleotide to be amplified, primer corresponding to said polynucleotide to be amplified, and/or a polynucleotide product to a bead may be via a photolabile linker, such as, for example, PC Amino C6.
  • a photolabile linker In cases where a photolabile linker is used, light may be used to release a linked polynucleotide from the bead.
  • the bead may be, for example, a magnetic bead or a latex bead.
  • the bead may then enable separation by, for example, magnetic sorting or sedimentation. Sedimentation of latex particles may be performed, for example, by centrifugation in a liquid that is more dense than latex, such as glycerol. In some cases, density gradient centrifugation may be used.
  • Beads may be of uniform size or heterogeneous size.
  • the diameter of a bead may be about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a bead may have a diameter of at least about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a bead may have a diameter of less than about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a bead may have a diameter of about 0.001 ⁇ m to 1 mm, 0.01 ⁇ m to 900 ⁇ m, 0.1 ⁇ m to 600 ⁇ m, 100 ⁇ m to 200 ⁇ m, 100 ⁇ m to 300 ⁇ m, 100 ⁇ m to 400 ⁇ m, 100 ⁇ m to 500 ⁇ m, 100 ⁇ m to 600 ⁇ m, 20 ⁇ m to 50 ⁇ m, 150 ⁇ m to 200 ⁇ m, 150 ⁇ m to 300 ⁇ m, or 150 ⁇ m to 400 ⁇ m.
  • a differential charge between the partitions comprising polynucleotides and partitions not comprising polynucleotides may be used to isolate partitions comprising polynucleotides, for example by performing electrophoresis or dielectrophoresis on the partitions.
  • partitions comprising polynucleotides may be isolated by flow fractionation, solvent extraction, differential melting (e.g., with nucleic acid probes), or freezing.
  • Isolation of partitions comprising polynucleotides provides a library of partitioned polynucleotide barcodes with significant diversity while incurring only a one-time bulk synthesis expense.
  • barcodes described in this disclosure can have a variety of structures.
  • barcodes of this disclosure are a part of an adapter.
  • an “adapter” is a structure used to enable attachment of a barcode to a target polynucleotide.
  • An adapter may comprise, for example, a barcode, polynucleotide sequence compatible for ligation with a target polynucleotide, and functional sequences such as primer binding sites and immobilization regions.
  • an adapter is a forked adapter.
  • An example of a forked adapter is schematically depicted in FIG. 1 .
  • two copies of a forked adapter structure 106 are depicted on opposite sides of a target polynucleotide 105 .
  • Each forked adapter comprises a first immobilization region 101 , a second immobilization region 102 , a first sequencing primer region 103 , a second sequencing primer region 104 and a pair of partially complementary regions (within 103 and 104 ) that anneal to each other.
  • Either the sequencing primer regions or immobilization regions may be used to immobilize the barcoded polynucleotides, for example, onto the surface of a bead.
  • the sequencing primer regions may be used, for example, as annealing sites for sequencing primers.
  • an overhang may be designed to enable compatibility with a target sequence.
  • the pair of annealed polynucleotides 103 and 104 have a 3′-T overhang, which is compatible with the 3′-A overhang on the target polynucleotide 105 .
  • a barcode may be included in any suitable portion of a forked adapter. After attachment of the forked adapter comprising the barcode to the target sequence 105 , the sequencing primer regions 103 and 104 can be used to sequence the target polynucleotide.
  • forked adapter structure includes those used in IlluminaTM library preparations and NEBNext® Multiplex Oligos for Illumina available from New England BiolabsTM.
  • non-forked adapters include those disclosed in Merriman et al., Electrophoresis, 33(23) 3397-3417 (2012), which is incorporated herein by reference, in its entirety.
  • FIG. 2 illustrates three schematic examples of placement of barcode regions within the forked adapter depicted in FIG. 1 .
  • a barcode 205 (BC1) is placed within the first immobilization region 201 or between the first immobilization region 201 and the first sequencing primer region 203 .
  • a barcode 206 (BC2) is placed within or adjacent to the first sequencing primer region 203 .
  • a barcode 207 (BC3) is placed within the second immobilization region 202 or between the second immobilization region 202 and the second sequencing primer region 204 .
  • FIG. 2 depicts barcodes on both ends of the target sequence, this is not necessary, as only one barcode per target sequence is sufficient for some applications. However, as described elsewhere in this disclosure, more than one barcode per target sequence may also be used.
  • FIG. 3 provides exemplary sequences (SEQ ID NO: 1 and SEQ ID NO: 22) of two forked adapters ligated to opposite ends of a target polynucleotide (NNN) and shows barcode regions of each forked adapter at the sequence level (bolded, nucleotides 30-37, 71-77, 81-87, and 122-129).
  • NNN target polynucleotide
  • nucleotides 1-29 represent an immobilization region of the first forked adapter
  • nucleotides 38-70 represent a sequencing primer region of the first forked adapter
  • nucleotides 78-80 represent a target polynucleotide of arbitrary length
  • nucleotides 88-120 represent a sequencing primer region of the second forked adapter
  • nucleotides 129-153 represent an immobilization region of the second forked adapter.
  • compositions, systems, devices, and kits of the disclosure may utilize the subdivision (partitioning) of certain species into separate partitions.
  • a partition may be, for example, a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a surface of a bead, or any other suitable container for sequestering one fraction of a sample or a species.
  • Partitions may be used to contain a species for further processing.
  • a species is a polynucleotide analyte
  • further processing may comprise cutting, ligating, and/or barcoding with species that are reagents.
  • Any number of devices, systems or containers may be used to hold, support or contain partitions.
  • a microwell plate may be used to hold, support, or contain partitions. Any suitable microwell plate may be used, for example microwell plates having 96, 384, or 1536 wells.
  • Each partition may also contain, or be contained within any other suitable partition.
  • a well, microwell, hole, a surface of a bead, or a tube may comprise a droplet (e.g., a droplet in an emulsion), a continuous phase in an emulsion, a spot, a capsule, or any other suitable partition.
  • a droplet may comprise a capsule, bead, or another droplet.
  • a capsule may comprise a droplet, bead, or another capsule.
  • a well or microwell comprises a plurality of droplets and a plurality of capsules.
  • a capsule comprises a plurality of capsules and a plurality of droplets. All combinations of partitions are envisioned. Table 1 shows non-limiting examples of partitions that may be combined with each other.
  • a partition may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions.
  • a partition may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, a partition may comprise less than 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, each partition may comprise 2-50, 2-20, 2-10, or 2-5 partitions.
  • a partition may comprise any suitable species or mixture of species.
  • a partition may comprise a reagent, an analyte, a sample, a cell, and combinations thereof.
  • a partition comprising other partitions may comprise certain species in the same partitions and certain species in different partitions. Species may be distributed between any suitable partitions, depending on the needs of the particular process.
  • any of the partitions in Table 1 may contain at least one first species and any of the partitions in Table 1 may contain at least one second species.
  • the first species may be a reagent and the second species may be an analyte.
  • a species is a polynucleotide isolated from a cell.
  • polynucleotides e.g., genomic DNA, RNA, etc.
  • the polynucleotide may be quantified.
  • the quantified polynucleotide may then be partitioned into a plurality of partitions as described herein.
  • the partitioning of the polynucleotide may be performed at a predetermined coverage amount, according to the quantification and the needs of the assay.
  • all or most (e.g., at least 50%, 60%, 70%, 80%, 90%, or 95%) of the partitions do not comprise polynucleotides that overlap, such that separate mixtures of non-overlapping fragments are formed across the plurality of partitions.
  • the partitioned polynucleotides may then be treated according to any suitable method known in the art or described in this disclosure.
  • the partitioned polynucleotides may be fragmented, amplified, barcoded, and the like.
  • Species may be partitioned using a variety of methods. For example, species may be diluted and dispensed across a plurality of partitions. A terminal dilution of a medium comprising species may be performed such that the number of partitions exceeds the number of species. Dilution may also be used prior to forming an emulsion or capsules, or prior to spotting a species on a substrate.
  • the ratio of the number of species to the number of partitions may be about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the ratio of the number of species to the number of partitions may be at least about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the ratio of the number of species to the number of partitions may be less than about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000.
  • the ratio of the number of species to the number of partitions may range from about 0.1-10, 0.5-10, 1-10, 2-10, 10-100, 100-1000, or more.
  • Partitioning may also be performed using piezoelectric droplet generation (e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520) or surface acoustic waves (e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145).
  • piezoelectric droplet generation e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520
  • surface acoustic waves e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145.
  • the number of partitions employed may vary depending on the application.
  • the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more.
  • the number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000.
  • the number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • the number of different barcodes or different sets of barcodes that are partitioned may vary depending upon, for example, the particular barcodes to be partitioned and/or the application. Different sets of barcodes may be, for example, sets of identical barcodes where the identical barcodes differ between each set. Or different sets of barcodes may be, for example, sets of different barcodes, where each set differs in its included barcodes.
  • less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes or different sets of barcodes may be partitioned. In some examples, about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes may be partitioned.
  • Barcodes may be partitioned at a particular density. For example, barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes per partition.
  • Barcodes may be partitioned such that identical barcodes are partitioned at a particular density.
  • identical barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more identical barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 identical barcodes per partition.
  • Barcodes may be partitioned such that different barcodes are partitioned at a particular density. For example, different barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition.
  • Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes per partition.
  • Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition.
  • Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 different barcodes per partition.
  • the number of partitions employed to partition barcodes may vary, for example, depending on the application and/or the number of different barcodes to be partitioned.
  • the number of partitions employed to partition barcodes may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more.
  • the number of partitions employed to partition barcodes may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more.
  • the number of partitions employed to partition barcodes may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, or 20000000.
  • the number of partitions employed to partition barcodes may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
  • different barcodes or different sets of barcodes may be partitioned such that each partition comprises a different barcode or different barcode set.
  • each partition may comprise a different set of identical barcodes.
  • the number of identical barcodes per partition may vary. For example, about 100,000 or more different sets of identical barcodes may be partitioned across about 100,000 or more different partitions, such that each partition comprises a different set of identical barcodes. In each partition, the number of identical barcodes per set of barcodes may be about 1,000,000 identical barcodes.
  • the number of different sets of barcodes may be equal to or substantially equal to the number of partitions. Any suitable number of different barcodes or different barcode sets (including numbers of different barcodes or different barcode sets to be partitioned described elsewhere herein), number of barcodes per partition (including numbers of barcodes per partition described elsewhere herein), and number of partitions (including numbers of partitions described elsewhere herein) may be combined to generate a diverse library of partitioned barcodes with high numbers of barcodes per partition.
  • any of the above-described different numbers of barcodes may be provided with any of the above-described barcode densities per partition, and in any of the above-described numbers of partitions.
  • the volume of the partitions may vary depending on the application.
  • the volume of any of the partitions described in this disclosure e.g., wells, spots, droplets (e.g., in an emulsion), and capsules
  • the volume of any of the partitions described in this disclosure may be about 1000 ⁇ l, 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 p
  • the volume of the partitions may be at least about 1000 ⁇ l, 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 pL, 300 pL, 200 pL, 100 pL, 50 pL, 25 pL, 10 pL, 5 pL, 1 pL, 900 fL, 800 pL, 700 fL
  • the volume of the partitions may be less than about 1000 ⁇ l, 900 ⁇ l, 800 ⁇ l, 700 ⁇ l, 600 ⁇ l, 500 ⁇ l, 400 ⁇ l, 300 ⁇ l, 200 ⁇ l, 100 ⁇ l, 50 ⁇ l, 25 ⁇ l, 10 ⁇ l, 5 ⁇ l, 1 ⁇ l, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 pL, 300 pL, 200 pL, 100 pL, 50 pL, 25 pL, 10 pL, 5 pL, 1 pL, 900 fL, 800 pL, 700 fL
  • volume of fluid in different partitions may vary by at least (or at most) plus or minus 1%, 2%, 3%, 4%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400%, 500%, or 1000% across a set of partitions.
  • a well or other partition
  • a capture reagent e.g., an oligonucleotide probe
  • a capture oligonucleotide may be immobilized or placed within a partition to capture specific species (e.g., polynucleotides).
  • a capture oligonucleotide may be immobilized on the surface of a bead in order to capture a species comprising an oligonucleotide with a complementary sequence.
  • Species may also be partitioned at a particular density. For example, species may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more species per partition. Species may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 species per partition.
  • Species may be partitioned such that at least one partition comprises a species that is unique within that partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for at least about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions.
  • wells are used as partitions.
  • the wells may be microwells.
  • a well may comprise a medium comprising a species or plurality of species. Species may be contained within a well in various configurations.
  • a species is dispensed directly into a well.
  • a species dispensed directly into a well may be overlaid with a layer that is, for example, dissolvable, meltable, or permeable. This layer may be, for example, an oil, wax, membrane, or the like.
  • the layer may be dissolved or melted prior to or after introduction of another species into the well.
  • the well may be sealed at any point, with a sealing layer, for example after addition of any species.
  • reagents for sample processing are dispensed directly into a well and overlaid with a layer that is dissolvable, meltable, or permeable.
  • a sample comprising an analyte to be processed is introduced on top of the layer.
  • the layer is dissolved or melted, or the analyte (or reagent) diffuses through the layer.
  • the well is sealed and incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered.
  • wells comprise other partitions.
  • a well may comprise any suitable partition including, for example, another well, a spot, a droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like.
  • Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • a well comprises a capsule comprising reagents for sample processing.
  • a capsule may be loaded into a well using a liquid medium, or loaded into a well without a liquid medium (e.g., essentially dry).
  • a capsule may contain one or more capsules, or other partitions.
  • a sample comprising an analyte to be processed may be introduced into the well.
  • the well may be sealed and a stimulus may be applied to cause release of the contents of the capsule into the well, resulting in contact between the reagents and the analyte to be processed.
  • the well may be incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the well, the opposite configuration—i.e., reagent in the well and analyte in the capsule—is also possible.
  • a well comprises an emulsion and the droplets of the emulsion comprise capsules comprising reagents for sample processing.
  • a sample comprising an analyte to be processed is contained within the droplets of the emulsion.
  • the well is sealed and a stimulus is applied to cause release of the contents of the capsules into the droplets, resulting in contact between the reagents and the analyte to be processed.
  • the well is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in a droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
  • Wells may be arranged as an array, for example a microwell array. Based on the dimensions of individual wells and the size of the substrate, the well array may comprise a range of well densities. In some cases, the well density may be 10 wells/cm 2 , 50 wells/cm 2 , 100 wells/cm 2 , 500 wells/cm 2 , 1000 wells/cm 2 , 5000 wells/cm 2 , 10000 wells/cm 2 , 50000 wells/cm 2 , or 100000 wells/cm 2 .
  • the well density may be at least 10 wells/cm 2 , 50 wells/cm 2 , 100 wells/cm 2 , 500 wells/cm 2 , 1000 wells/cm 2 , 5000 wells/cm 2 , 10000 wells/cm 2 , 50000 wells/cm 2 , or 100000 wells/cm 2 .
  • the well density may be less than 10 wells/cm 2 , 50 wells/cm 2 , 100 wells/cm 2 , 500 wells/cm 2 , 1000 wells/cm 2 , 5000 wells/cm 2 , 10000 wells/cm 2 , 50000 wells/cm 2 , or 100000 wells/cm 2 .
  • spots are used as partitions.
  • a spot may be made, for example, by dispensing a substance on a surface. Species may be contained within a spot in various configurations.
  • a species is dispensed directly into a spot by including the species in the medium used to form the spot.
  • a species dispensed directly onto a spot may be overlaid with a layer that is, for example, dissolvable, meltable, or permeable. This layer may be, for example, an oil, wax, membrane, or the like. The layer may be dissolved or melted prior to or after introduction of another species onto the spot.
  • the spot may be sealed at any point, for example after addition of any species, by an overlay.
  • reagents for sample processing are dispensed directly onto a spot, for example on a glass slide, and overlaid with a layer that is dissolvable, meltable, or permeable.
  • a sample comprising an analyte to be processed is introduced on top of the layer.
  • the layer is dissolved or melted, or the analyte (or reagent) diffuses through the layer.
  • the spot is sealed and incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered.
  • spots may also be arranged within a well.
  • a plurality of spots may be arranged within a well such that the contents of each spot do not mix.
  • Such a configuration may be useful, for example, when it is desirable to prevent species from contacting each other.
  • a well may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more spots.
  • a well may comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more spots.
  • a well may comprise less than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 spots.
  • a well may comprise 2-4, 2-6, 2-8, 4-6, 4-8, 5-10, or 4-12 spots.
  • a substance e.g., a medium containing an analyte
  • the species in the spot may mix.
  • using separate spots to contain different species (or combinations of species) may also be useful to prevent cross-contamination of devices used to place the spots inside the well.
  • spots comprise other partitions.
  • a spot may comprise any suitable partition including, for example, another spot a droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like.
  • Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • a spot comprises a capsule comprising reagents for sample processing.
  • a capsule may contain one or more capsules, or other partitions.
  • a sample comprising an analyte to be processed is introduced into the spot.
  • the spot is sealed and a stimulus is applied to cause release of the contents of the capsule into the spot, resulting in contact between the reagents and the analyte to be processed.
  • the spot is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the spot, the opposite configuration—i.e., reagent in the spot and analyte in the capsule—is also possible.
  • a spot comprises an emulsion and the droplets of the emulsion comprise capsules comprising reagents for sample processing.
  • a sample comprising an analyte to be processed is contained within the droplets of the emulsion.
  • the spot is sealed and a stimulus is applied to cause release of the contents of the capsules into the droplets, resulting in contact between the reagents and the analyte to be processed.
  • the spot is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in a droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
  • Spots may be of uniform size or heterogeneous size.
  • the diameter of a spot may be about 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, 1 mm, 2 mm, 5 mm, or 1 cm.
  • a spot may have a diameter of at least about 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, 1 mm, 1 mm, 2 mm, 5 mm, or 1 cm.
  • a spot may have a diameter of less than about 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, 1 mm, 1 mm, 2 mm, 5 mm, or 1 cm.
  • a spot may have a diameter of about 0.1 ⁇ m to 1 cm, 100 ⁇ m to 1 mm, 100 ⁇ m to 500 ⁇ m, 100 ⁇ m to 600 ⁇ m, 150 ⁇ m to 300 ⁇ m, or 150 ⁇ m to 400 ⁇ m.
  • Spots may be arranged as an array, for example a spot array. Based on the dimensions of individual spots and the size of the substrate, the spot array may comprise a range of spot densities. In some cases, the spot density may be 10 spots/cm 2 , 50 spots/cm 2 , 100 spots/cm 2 , 500 spots/cm 2 , 1000 spots/cm 2 , 5000 spots/cm 2 , 10000 spots/cm 2 , 50000 spots/cm 2 , or 100000 spots/cm 2 .
  • the spot density may be at least 10 spots/cm 2 , 50 spots/cm 2 , 100 spots/cm 2 , 500 spots/cm 2 , 1000 spots/cm 2 , 5000 spots/cm 2 , 10000 spots/cm 2 , 50000 spots/cm 2 , or 100000 spots/cm 2 . In some cases, the spot density may be less than 10 spots/cm 2 , 50 spots/cm 2 , 100 spots/cm 2 , 500 spots/cm 2 , 1000 spots/cm 2 , 5000 spots/cm 2 , 10000 spots/cm 2 , 50000 spots/cm 2 , or 100000 spots/cm 2 .
  • the droplets in an emulsion are used as partitions.
  • An emulsion may be prepared, for example, by any suitable method, including methods known in the art. (See e.g., Weizmann et al., Nature Methods, 2006, 3(7):545-550; Weitz et al. U.S. Pub. No. 2012/0211084).
  • water-in-fluorocarbon emulsions may be used. These emulsions may incorporate fluorosurfactants such as oligomeric perfluorinated polyethers (PFPE) with polyethylene glycol (PEG). (Holtze et al., Lab on a Chip, 2008, 8(10):1632-1639).
  • PFPE oligomeric perfluorinated polyethers
  • PEG polyethylene glycol
  • monodisperse emulsions may be formed in a microfluidic flow focusing device. (Garstecki et al., Applied Physics Letters, 2004, 85(13
  • a species may be contained within a droplet in an emulsion containing, for example, a first phase (e.g., oil or water) forming the droplet and a second (continuous) phase (e.g., water or oil).
  • a first phase e.g., oil or water
  • a second phase e.g., water or oil
  • An emulsion may be a single emulsion, for example, a water-in-oil or an oil-in-water emulsion.
  • An emulsion may be a double emulsion, for example a water-in-oil-in-water or an oil-in-water-in-oil emulsion. Higher-order emulsions are also possible.
  • the emulsion may be held in any suitable container, including any suitable partition described in this disclosure.
  • droplets in an emulsion comprise other partitions.
  • a droplet in an emulsion may comprise any suitable partition including, for example, another droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like.
  • Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • a droplet in an emulsion comprises a capsule comprising reagents for sample processing.
  • a capsule may contain one or more capsules, or other partitions.
  • a sample comprising an analyte to be processed is contained within the droplet.
  • a stimulus is applied to cause release of the contents of the capsule into the droplet, resulting in contact between the reagents and the analyte to be processed.
  • the droplet is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
  • the droplets in an emulsion may be of uniform size or heterogeneous size.
  • the diameter of a droplet in an emulsion may be about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of at least about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of less than about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a droplet may have a diameter of about 0.001 ⁇ m to 1 mm, 0.01 ⁇ m to 900 ⁇ m, 0.1 ⁇ m to 600 ⁇ m, 100 ⁇ m to 200 ⁇ m, 100 ⁇ m to 300 ⁇ m, 100 ⁇ m to 400 ⁇ m, 100 ⁇ m to 500 ⁇ m, 100 ⁇ m to 600 ⁇ m, 150 ⁇ m to 200 ⁇ m, 150 ⁇ m to 300 ⁇ m, or 150 ⁇ m to 400 ⁇ m.
  • Droplets in an emulsion also may have a particular density.
  • the droplets are less dense than an aqueous fluid (e.g., water); in some cases, the droplets are denser than an aqueous fluid.
  • the droplets are less dense than a non-aqueous fluid (e.g., oil); in some cases, the droplets are denser than a non-aqueous fluid.
  • Droplets may have a density of about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g/
  • Droplets may have a density of at least about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96
  • droplet densities may be at most about 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g/cm 3 , 0.97 g/cm 3 , 0.98 g/cm 3 , 0.99 g/cm 3 , 1.00 g/cm 3 , 1.05 g/cm 3 , 1.1 g/cm 3 ,
  • capsules are used as partitions.
  • a capsule may be prepared by any suitable method, including methods known in the art, including emulsification polymerization (Weitz et al. (U.S. Pub. No. 2012/0211084)), layer-by-layer assembly with polyelectrolytes, coacervation, internal phase separation, and flow focusing. Any suitable species may be contained within a capsule.
  • the capsule may be held in any suitable container, including any suitable partition described in this disclosure.
  • capsules comprise other partitions.
  • a capsule may comprise any suitable partition including, for example, another capsule, a droplet in an emulsion, a bead, and the like.
  • Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
  • an outer capsule comprises an inner capsule.
  • the inner capsule comprises reagents for sample processing.
  • An analyte is encapsulated in the medium between the inner capsule and the outer capsule.
  • a stimulus is applied to cause release of the contents of the inner capsule into the outer capsule, resulting in contact between the reagents and the analyte to be processed.
  • the outer capsule is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered.
  • Capsules may be pre-formed and filled with reagents by injection.
  • the picoinjection methods described in Abate et al. Proc. Natl. Acad. Sci. U.S.A., 2010, 107(45), 19163-19166) and Weitz et al. (U.S. Pub. No. 2012/0132288) may be used to introduce reagents into the interior of capsules described herein.
  • the picoinjection will be performed prior to the hardening of the capsule shell, for example by injecting species into the interior of a capsule precursor, such as a droplet of an emulsion, before formation of the capsule shell.
  • Capsules may be of uniform size or heterogeneous size.
  • the diameter of a capsule may be about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a capsule may have a diameter of at least about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a capsule may have a diameter of less than about 0.001 ⁇ m, 0.01 ⁇ m, 0.05 ⁇ m, 0.1 ⁇ m, 0.5 ⁇ m, 1 ⁇ m, 5 ⁇ m, 10 ⁇ m, 50 ⁇ m, 100 ⁇ m, 150 ⁇ m, 200 ⁇ m, 300 ⁇ m, 400 ⁇ m, 500 ⁇ m, 600 ⁇ m, 700 ⁇ m, 800 ⁇ m, 900 ⁇ m, or 1 mm.
  • a capsule may have a diameter of about 0.001 ⁇ m to 1 mm, 0.01 ⁇ m to 900 ⁇ m, 0.1 ⁇ m to 600 ⁇ m, 100 ⁇ m to 200 ⁇ m, 100 ⁇ m to 300 ⁇ m, 100 ⁇ m to 400 ⁇ m, 100 ⁇ m to 500 ⁇ m, 100 ⁇ m to 600 ⁇ m, 150 ⁇ m to 200 ⁇ m, 150 ⁇ m to 300 ⁇ m, or 150 ⁇ m to 400 ⁇ m.
  • Capsules also may have a particular density.
  • the capsules are less dense than an aqueous fluid (e.g., water); in some cases, the capsules are denser than an aqueous fluid.
  • the capsules are less dense than a non-aqueous fluid (e.g., oil); in some cases, the capsules are denser than a non-aqueous fluid.
  • Capsules may have a density of about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96
  • Capsules may have a density of at least about 0.05 g/cm 3 , 0.1 g/cm 3 , 0.2 g/cm 3 , 0.3 g/cm 3 , 0.4 g/cm 3 , 0.5 g/cm 3 , 0.6 g/cm 3 , 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 ,
  • capsule densities may be at most about 0.7 g/cm 3 , 0.8 g/cm 3 , 0.81 g/cm 3 , 0.82 g/cm 3 , 0.83 g/cm 3 , 0.84 g/cm 3 , 0.85 g/cm 3 , 0.86 g/cm 3 , 0.87 g/cm 3 , 0.88 g/cm 3 , 0.89 g/cm 3 , 0.90 g/cm 3 , 0.91 g/cm 3 , 0.92 g/cm 3 , 0.93 g/cm 3 , 0.94 g/cm 3 , 0.95 g/cm 3 , 0.96 g/cm 3 , 0.97 g/cm 3 , 0.98 g/cm 3 , 0.99 g/cm 3 , 1.00 g/cm 3 , 1.05 g/cm 3 , 1.1 g/cm 3 , 1.2
  • capsules may be produced by flow focusing.
  • Flow focusing is a method whereby a first fluid that is immiscible with a second fluid is flowed into the second fluid.
  • a first (e.g., aqueous) fluid comprising a monomer, crosslinker, initiator, and aqueous surfactant 1201 is flowed into a second (e.g., oil) fluid comprising a surfactant and an accelerator 1202 .
  • a droplet of first fluid breaks off from the first fluid stream and a capsule shell begins to form 1204 due to the mixing of the monomer, crosslinker, and initiator in the first fluid and the accelerator in the second fluid.
  • a capsule is formed.
  • the shell becomes thicker due to increased exposure to the accelerator. Varying the concentrations of the reagents may also be used to vary the thickness and permeability of the capsule shell.
  • a species, or other partition such as a droplet may be encapsulated by, for example, including the species in the first fluid. Including the species in the second fluid may embed the species in the shell of the capsule.
  • the phases may also be reversed—i.e., the first phase may be an oil phase and the second phase may be an aqueous phase.
  • capsules within capsules may be produced by flow focusing.
  • a first (e.g., aqueous) fluid comprising a capsule, monomer, crosslinker, initiator, and aqueous surfactant 1301 is flowed into a second (oil) fluid comprising a surfactant and an accelerator 1302 .
  • a droplet of first fluid breaks off from the first fluid stream and a second capsule shell begins to form around the capsule 1304 due to the mixing of the monomer, crosslinker, and initiator in the first fluid and the accelerator in the second fluid.
  • a capsule within a capsule is formed.
  • the shell becomes thicker due to increased exposure to the accelerator. Varying the concentrations of the reagents may also be used to vary the thickness and permeability of the second capsule shell.
  • a species may be encapsulated by, for example, including the species in the first fluid. Including the species in the second fluid may embed the species in the second shell of the capsule.
  • the phases may also be reversed—i.e., the first phase may be an oil phase and the second phase may be an aqueous phase.
  • capsules may be produced in batch, using capsule precursors, such as the droplets in an emulsion.
  • Capsule precursors may be formed by any suitable method, for example by producing an emulsion with droplets comprising a monomer, a crosslinker, an initiator, and a surfactant.
  • An accelerator may then be added to the medium, resulting in the formation of capsules.
  • the thickness of the shell can be varied by varying the concentrations of the reactants, and the time of exposure to the accelerator.
  • the capsules may then be washed and recovered.
  • a species, including other partitions may be encapsulated within the capsule or, if suitable, within the shell.
  • the droplets of an emulsion may be exposed to an accelerator that is present in an outlet well during the emulsion generation process.
  • capsule precursors may be formed by any suitable method, such as the flow focusing method illustrated in FIG. 12 .
  • the accelerator may be included in a medium located at the exit of the T-junction (e.g., a medium located at the far-right of the horizontal channel of FIG. 12 .
  • the emulsion droplets i.e., capsule precursors
  • the medium comprising the accelerator i.e., the outlet medium.
  • the capsule precursor has a density that is less than the density of outlet medium, the capsule precursors will rise through the medium, ensuring convectional and diffusional exposure to the accelerator and reducing the likelihood of polymerization at the outlet of the channel.
  • a species can be, for example, any substance used in sample processing, such as a reagent or an analyte.
  • Exemplary species include whole cells, chromosomes, polynucleotides, organic molecules, proteins, polypeptides, carbohydrates, saccharides, sugars, lipids, enzymes, restriction enzymes, ligases, polymerases, barcodes, adapters, small molecules, antibodies, fluorophores, deoxynucleotide triphosphates (dNTPs), dideoxynucleotide triphosphates (ddNTPs), buffers, acidic solutions, basic solutions, temperature-sensitive enzymes, pH-sensitive enzymes, light-sensitive enzymes, metals, metal ions, magnesium chloride, sodium chloride, manganese, aqueous buffer, mild buffer, ionic buffer, inhibitors, saccharides, oils, salts, ions, detergents, ionic detergents, non-
  • a partition comprises a set of species that have a similar attribute (e.g., a set of enzymes, a set of minerals, a set of oligonucleotides, a mixture of different barcodes, a mixture of identical barcodes).
  • a partition comprises a heterogeneous mixture of species.
  • the heterogeneous mixture of species comprises all components necessary to perform a particular reaction.
  • such mixture comprises all components necessary to perform a reaction, except for 1, 2, 3, 4, 5, or more components necessary to perform the reaction.
  • additional components are contained within a different partition or within a solution within or surrounding a partition.
  • a species may be naturally-occurring or synthetic.
  • a species may be present in a sample obtained using any methods known in the art.
  • a sample may be processed before analyzing it for an analyte.
  • a species may be obtained from any suitable location, including from organisms, whole cells, cell preparations and cell-free compositions from any organism, tissue, cell, or environment.
  • a species may be obtained from environmental samples, biopsies, aspirates, formalin fixed embedded tissues, air, agricultural samples, soil samples, petroleum samples, water samples, or dust samples.
  • a species may be obtained from bodily fluids which may include blood, urine, feces, serum, lymph, saliva, mucosal secretions, perspiration, central nervous system fluid, vaginal fluid, or semen.
  • Species may also be obtained from manufactured products, such as cosmetics, foods, personal care products, and the like.
  • Species may be the products of experimental manipulation including, recombinant cloning, polynucleotide amplification, polymerase chain reaction (PCR) amplification, purification methods (such as purification of genomic DNA or RNA), and synthesis reactions.
  • PCR polymerase chain reaction
  • a species may quantified by mass.
  • a species may be provided in a mass of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng, 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 15 ⁇ g, or 20 ⁇ g.
  • a species may be provided in a mass of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng, 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 15 ⁇ g, or 20 ⁇ g.
  • a species may be provided in a mass of less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 15 ⁇ g, or 20 ⁇ g.
  • a species may be provided in a mass ranging from about 1-10, 10-50, 50-100, 100-200, 200-1000, 1000-10000 ng, 1-5 ⁇ g, or 1-20 ⁇ g.
  • amplification may be used to increase the quantity of a polynucleotide.
  • Polynucleotides may also be quantified as “genome equivalents.”
  • a genome equivalent is an amount of polynucleotide equivalent to one haploid genome of an organism from which the target polynucleotide is derived. For example, a single diploid cell contains two genome equivalents of DNA.
  • Polynucleotides may be provided in an amount ranging from about 1-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 genome equivalents.
  • Polynucleotides may be provided in an amount of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
  • Polynucleotides may be provided in an amount less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
  • Polynucleotides may also be quantified by the amount of sequence coverage provided.
  • the amount of sequence coverage refers to the average number of reads representing a given nucleotide in a reconstructed sequence. Generally, the greater the number of times a region is sequenced, the more accurate the sequence information obtained.
  • Polynucleotides may be provided in an amount that provides a range of sequence coverage from about 0.1 ⁇ -10 ⁇ , 10 ⁇ -50 ⁇ , 50 ⁇ -100 ⁇ , 100 ⁇ -200 ⁇ , or 200 ⁇ -500 ⁇ .
  • Polynucleotides may be provided in an amount that provides at least about 0.1 ⁇ , 0.2 ⁇ , 0.3 ⁇ , 0.4 ⁇ , 0.5 ⁇ , 0.6 ⁇ , 0.7 ⁇ , 0.8 ⁇ , 0.9 ⁇ , 1.0 ⁇ , 5 ⁇ , 10 ⁇ , 25 ⁇ , 50 ⁇ , 100 ⁇ , 125 ⁇ , 150 ⁇ , 175 ⁇ , or 200 ⁇ sequence coverage. Polynucleotides may be provided in an amount that provides less than about 0.2 ⁇ , 0.3 ⁇ , 0.4 ⁇ , 0.5 ⁇ , 0.6 ⁇ , 0.7 ⁇ , 0.8 ⁇ , 0.9 ⁇ , 1.0 ⁇ , 5 ⁇ , 10 ⁇ , 25 ⁇ , 50 ⁇ , 100 ⁇ , 125 ⁇ , 150 ⁇ , 175 ⁇ , or 200 ⁇ sequence coverage.
  • species are introduced into a partition either before or after a particular step.
  • a lysis buffer reagent may be introduced into a partition following partitioning of a cellular sample into the partitions.
  • reagents and/or partitions comprising reagents are introduced sequentially such that different reactions or operations occur at different steps.
  • the reagents (or partitions comprising reagents) may be also be loaded at steps interspersed with a reaction or operation step.
  • capsules comprising reagents for fragmenting molecules may be loaded into a well, followed by a fragmentation step, which may be followed by loading of capsules comprising reagents for ligating barcodes (or other unique identifiers, e.g., antibodies) and subsequent ligation of the barcodes to the fragmented molecules.
  • reagents for fragmenting molecules e.g., nucleic acids
  • a fragmentation step which may be followed by loading of capsules comprising reagents for ligating barcodes (or other unique identifiers, e.g., antibodies) and subsequent ligation of the barcodes to the fragmented molecules.
  • the methods, compositions, systems, devices, and kits of this disclosure may be used to process a sample containing a species, for example an analyte. Any suitable process can be performed.
  • the methods, compositions, systems, devices, and kits of this disclosure may be used for polynucleotide fragmentation. Fragmentation of polynucleotides is used as a step in a variety of methods, including polynucleotide sequencing.
  • the size of the polynucleotide fragments typically described in terms of length (quantified by the linear number of nucleotides per fragment), may vary depending on the source of the target polynucleotide, the method used for fragmentation, and the desired application. A single fragmentation step or a plurality of fragmentation steps may be used.
  • Fragments generated using the methods described herein may be about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides in length. Fragments generated using the methods described herein may be at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides in length. Fragments generated using the methods described herein may be less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides in length.
  • Fragments generated using the methods described herein may have a mean or median length of about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides.
  • Fragments generated using the methods described herein may have a mean or median length of at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides. Fragments generated using the methods described herein may have a mean or median length of less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides.
  • fragmentation may be performed through physical, mechanical or enzymatic methods.
  • Physical fragmentation may include exposing a target polynucleotide to heat or to UV light.
  • Mechanical disruption may be used to mechanically shear a target polynucleotide into fragments of the desired range.
  • Mechanical shearing may be accomplished through a number of methods known in the art, including repetitive pipetting of the target polynucleotide, sonication (e.g., using ultrasonic waves), cavitation and nebulization.
  • Target polynucleotides may also be fragmented using enzymatic methods. In some cases, enzymatic digestion may be performed using enzymes such as using restriction enzymes.
  • this polynucleotide may be a fragment of a target polynucleotide which one wishes to further fragment. In still other cases, still further fragments may be still further fragmented. Any suitable polynucleotide may be fragmented according the methods described herein.
  • Restriction enzymes may be used to perform specific or non-specific fragmentation of target polynucleotides.
  • the methods of the present disclosure may use one or more types of restriction enzymes, generally described as Type I enzymes, Type II enzymes, and/or Type III enzymes.
  • Type II and Type III enzymes are generally commercially available and well known in the art.
  • Type II and Type III enzymes recognize specific sequences of nucleotide base pairs within a double stranded polynucleotide sequence (a “recognition sequence” or “recognition site”). Upon binding and recognition of these sequences, Type II and Type III enzymes cleave the polynucleotide sequence.
  • cleavage will result in a polynucleotide fragment with a portion of overhanging single stranded DNA, called a “sticky end.” In other cases, cleavage will not result in a fragment with an overhang, creating a “blunt end.”
  • the methods of the present disclosure may comprise use of restriction enzymes that generate either sticky ends or blunt ends.
  • Restriction enzymes may recognize a variety of recognition sites in the target polynucleotide. Some restriction enzymes (“exact cutters”) recognize only a single recognition site (e.g., GAATTC). Other restriction enzymes are more promiscuous, and recognize more than one recognition site, or a variety of recognition sites. Some enzymes cut at a single position within the recognition site, while others may cut at multiple positions. Some enzymes cut at the same position within the recognition site, while others cut at variable positions.
  • the present disclosure provides method of selecting one or more restriction enzymes to produce fragments of a desired length.
  • Polynucleotide fragmentation may be simulated in silico, and the fragmentation may be optimized to obtain the greatest number or fraction of polynucleotide fragments within a particular size range, while minimizing the number or fraction of fragments within undesirable size ranges. Optimization algorithms may be applied to select a combination of two or more enzymes to produce the desired fragment sizes with the desired distribution of fragments quantities.
  • a polynucleotide may be exposed to two or more restriction enzymes simultaneously or sequentially. This may be accomplished by, for example, adding more than one restriction enzyme to a partition, or by adding one restriction enzyme to a partition, performing the digestion, deactivating the restriction enzyme (e.g., by heat treatment) and then adding a second restriction enzyme. Any suitable restriction enzyme may be used alone, or in combination, in the methods presented herein.
  • a species is a restriction enzyme that is a “rare-cutter.”
  • the term “rare-cutter enzyme,” as used herein, generally refers to an enzyme with a recognition site that occurs only rarely in a genome.
  • the size of restriction fragments generated by cutting a hypothetical random genome with a restriction enzyme may be approximated by 4 N , where N is the number of nucleotides in the recognition site of the enzyme. For example, an enzyme with a recognition site consisting of 7 nucleotides would cut a genome once every 4 7 bp, producing fragments of about 16,384 bp.
  • rare-cutter enzymes have recognition sites comprising 6 or more nucleotides.
  • a rare cutter enzyme may have a recognition site comprising or consisting of 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 nucleotides.
  • rare-cutter enzymes include NotI (GCGGCCGC), XmaIII (CGGCCG), SstII (CCGCGG), SalI (GTCGAC), NruI (TCGCGA), NheI (GCTAGC), Nb.BbvCI (CCTCAGC), BbvCI (CCTCAGC), AscI (GGCGCGCC), AsiSI (GCGATCGC), FseI (GGCCGGCC), PacI (TTAATTAA), PmeI (GTTTAAAC), SbfI (CCTGCAGG), SgrAI (CRCCGGYG), SwaI (ATTTAAAT), BspQI (GCTCTTC), SapI (GCTCTTC), SfiI (GGCCNNNNNGGCC) (SEQ ID NO: 27), CspCI (CAANNNNNGTGG) (SEQ ID NO: 28),
  • polynucleotides may be fragmented and barcoded at the same time.
  • a transposase e.g., NEXTERA
  • NEXTERA e.g., NEXTERA
  • stimuli may be used to trigger the release of a species from a partition.
  • a stimulus may cause disruption of the structure of a partition, such as the wall of a well, a component of a spot, the stability of a droplet (e.g., a droplet in an emulsion), or the shell of a capsule.
  • These stimuli are particularly useful in inducing a partition to release its contents.
  • stimuli-responsiveness may be employed to release the contents of one partition (e.g., a partition responsive to the stimulus) into another partition (e.g., a partition not responsive to that stimulus, or less responsive to that stimulus).
  • the contents of an inner capsule may be released into the contents of an outer capsule by applying a stimulus that dissolves the inner capsule, resulting in a capsule containing a mixed sample.
  • a stimulus that dissolves the inner capsule resulting in a capsule containing a mixed sample.
  • this embodiment is purely illustrative, and stimuli-responsiveness may be used to release the contents of any suitable partition into any other suitable partition, medium, or container (see, e.g., Table 1 for more specific examples of partitions within partitions).
  • stimuli examples include chemical stimuli, bulk changes, biological stimuli, light, thermal stimuli, magnetic stimuli, addition of a medium to a well, and any combination thereof, as described more fully below. (See, e.g., Esser-Kahn et al., (2011) Macromolecules 44: 5539-5553; Wang et al., (2009) ChemPhysChem 10:2405-2409.)
  • Numerous chemical triggers may be used to trigger the disruption of partitions (e.g., Plunkett et al., Biomacromolecules, 2005, 6:632-637).
  • Examples of these chemical changes may include, but are not limited to pH-mediated changes to the integrity of a component of a partition, disintegration of a component of a partition via chemical cleavage of crosslink bonds, and triggered depolymerization of a component of a partition.
  • Bulk changes may also be used to trigger disruption of partitions.
  • a change in pH of a solution may trigger disruption of a partition via a number of different mechanisms.
  • the addition of acid may cause degradation or disassembly a portion of a partition through a variety of mechanisms.
  • Addition of protons may disassemble cross-linking of polymers in a component of a partition, disrupt ionic or hydrogen bonds in a component of a partition, or create nanopores in a component of a partition to allow the inner contents to leak through to the exterior.
  • a change in pH may also destabilize an emulsion, leading to release of the contents of the droplets.
  • a partition is produced from materials that comprise acid-degradable chemical cross-linkers, such a ketals.
  • a decrease in pH, particular to a pH lower than 5, may induce the ketal to convert to a ketone and two alcohols and facilitate disruption of the partition.
  • the partitions may be produced from materials comprising one or more polyelectrolytes that are pH sensitive. A decrease in pH may disrupt the ionic- or hydrogen-bonding interactions of such partitions, or create nanopores therein.
  • partitions made from materials comprising polyelectrolytes comprise a charged, gel-based core that expands and contracts upon a change of pH.
  • Disruption of cross-linked materials comprising a partition can be accomplished through a number of mechanisms.
  • a partition can be contacted with various chemicals that induce oxidation, reduction or other chemical changes.
  • a reducing agent such as beta-mercaptoethanol
  • enzymes may be added to cleave peptide bonds in materials forming a partition, thereby resulting in a loss of integrity of the partition.
  • Depolymerization can also be used to disrupt partitions.
  • a chemical trigger may be added to facilitate the removal of a protecting head group.
  • the trigger may cause removal of a head group of a carbonate ester or carbamate within a polymer, which in turn causes depolymerization and release of species from the inside of a partition.
  • a chemical trigger may comprise an osmotic trigger, whereby a change in ion or solute concentration in a solution induces swelling of a material used to make a partition. Swelling may cause a buildup of internal pressure such that a partition ruptures to release its contents. Swelling may also cause an increase in the pore size of the material, allowing species contained within the partition to diffuse out, and vice versa.
  • a partition may also be made to release its contents via bulk or physical changes, such as pressure induced rupture, melting, or changes in porosity.
  • Biological stimuli may also be used to trigger disruption of partitions.
  • biological triggers resemble chemical triggers, but many examples use biomolecules, or molecules commonly found in living systems such as enzymes, peptides, saccharides, fatty acids, nucleic acids and the like.
  • partitions may be made from materials comprising polymers with peptide cross-links that are sensitive to cleavage by specific proteases. More specifically, one example may comprise a partition made from materials comprising GFLGK (SEQ ID NO: 34) peptide cross links.
  • GFLGK SEQ ID NO: 34
  • partitions comprise a component comprising cellulose. Addition of the hydrolytic enzyme chitosan serves as biologic trigger for cleavage of cellulosic bonds, depolymerization of component of the partition comprising chitosan, and release of its inner contents.
  • Partitions may also be induced to release their contents upon the application of a thermal stimulus.
  • a change in temperature can cause a variety changes to a partition.
  • a change in heat may cause melting of a partition such that a portion of the partition disintegrates, or disruption of an emulsion.
  • heat may increase the internal pressure of the inner components of a partition such that the partition ruptures or explodes.
  • heat may transform a partition into a shrunken dehydrated state. Heat may also act upon heat-sensitive polymers used as materials to construct partitions.
  • a partition is made from materials comprising a thermo-sensitive hydrogel.
  • a thermo-sensitive hydrogel Upon the application of heat, such as a temperature above 35 C, the hydrogel material shrinks. The sudden shrinkage of the material increases the pressure and ruptures the partition.
  • a material used to produce a partition may comprise a diblock polymer, or a mixture of two polymers, with different heat sensitivities.
  • One polymer may be particularly likely to shrink after the application of heat, while the other is more heat-stable. When heat is applied to such shell wall, the heat-sensitive polymer may shrink, while the other remains intact, causing a pore to form.
  • a material used to produce a partition may comprise magnetic nanoparticles. Exposure to a magnetic field may cause the generation of heat, leading to rupture of the partition.
  • Inclusion of magnetic nanoparticles in a material used to produce a partition may allow triggered rupture of the partition, as described above, as well as enable guidance of these partitions to other partitions (e.g., guidance of capsules to wells in an array).
  • incorporation of Fe 3 O 4 nanoparticles into materials used to produce partitions triggers rupture in the presence of an oscillating magnetic field stimulus.
  • a partition may also be disrupted as the result of electrical stimulation. Similar to the magnetic particles described in the previous section, electrically sensitive particles can allow for both triggered rupture of partitions, as well as other functions such as alignment in an electric field or redox reactions. In one example, partitions made from materials comprising electrically sensitive material are aligned in an electric field such that release of inner reagents can be controlled. In other examples, electric fields may induce redox reactions within a partition that may increase porosity.
  • a light stimulus may also be used to disrupt the partitions.
  • Numerous light triggers are possible and may include systems that use various molecules such as nanoparticles and chromophores capable of absorbing photons of specific ranges of wavelengths.
  • metal oxide coatings can be used to produce certain partitions. UV irradiation of partitions coated with SiO2/TiO2 may result in disintegration of the partition wall.
  • photo switchable materials such as azobenzene groups may be incorporated in the materials used to produce the partitions. Upon the application of UV or visible light, chemicals such as these undergo a reversible cis-to-trans isomerization upon absorption of photons. In this aspect, incorporation of photo switches results in disintegration of a portion of a partition, or an increase in porosity of a portion of a partition.
  • the devices, methods, compositions, systems, and kits of this disclosure may be used in combination with any apparatus or device that provides such trigger or stimulus.
  • a device may be used in combination with a heated or thermally controlled plate, which allows heating of the wells and may induce the rupture of capsules.
  • Any of a number of methods of heat transfer may be used for thermal stimuli, including but not limited to applying heat by radiative heat transfer, convective heat transfer, or conductive heat transfer.
  • the stimulus is a biological enzyme
  • the enzyme may be injected into a device such that it is deposited into each well.
  • a device may be used in combination with a magnetic or electric plate.
  • Sequencing may be performed by any available technique. For example, sequencing may be performed by the classic Sanger sequencing method. Sequencing methods may also include: high-throughput sequencing, pyrosequencing, sequencing-by-synthesis, single-molecule sequencing, nanopore sequencing, sequencing-by-ligation, sequencing-by-hybridization, RNA-Seq (Illumina), Digital Gene Expression (Helicos), next generation sequencing, single molecule sequencing by synthesis (SMSS) (Helicos), massively-parallel sequencing, clonal single molecule Array (Solexa), shotgun sequencing, Maxim-Gilbert sequencing, primer walking, and any other sequencing methods known in the art.
  • varying numbers of fragments are sequenced. For example, in some cases about 30%-90% of the fragments are sequenced. In some cases, about 35%-85%, 40%-80%, 45%-75%, 50%-70%, 55%-65%, or 50%-60% of the fragments are sequenced. In some cases, at least about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced. In some cases less than about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced.
  • sequences from fragments are assembled to provide sequence information for a contiguous region of the original target polynucleotide that is longer than the individual sequence reads.
  • Individual sequence reads may be about 10-50, 50-100, 100-200, 200-300, 300-400, or more nucleotides in length.
  • the identities of the barcode tags may serve to order the sequence reads from individual fragments as well as to differentiate between haplotypes. For example, during the partitioning of individual fragments, parental polynucleotide fragments may separated into different partitions. With an increase in the number of partitions, the likelihood of a fragment from both a maternal and paternal haplotype contained in the same partition becomes negligibly small. Thus, sequence reads from fragments in the same partition may be assembled and ordered.
  • This disclosure also provides methods and compositions to prepare polynucleotide fragments in such a manner that may enable phasing or linkage information to be generated. Such information may allow for the detection of linked genetic variations in sequences, including genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) that are separated by long stretches of polynucleotides.
  • the term “indel” refers to a mutation resulting in a colocalized insertion and deletion and a net gain or loss in nucleotides.
  • a “microindel” is an indel that results in a net gain or loss of 1 to 50 nucleotides.
  • These variations may exist in either a cis or trans relationship. In a cis relationship, two or more genetic variations exist in the same polynucleotide or strand. In a trans relationship, two or more genetic variations exist on multiple polynucleotide molecules or strands.
  • Methods provided herein may be used to determine polynucleotide phasing.
  • a polynucleotide sample e.g., a polynucleotide that spans a given locus or loci
  • the polynucleotide may then be fragmented, barcoded, and sequenced.
  • the sequences may be examined for genetic variation.
  • the detection of genetic variations in the same sequence tagged with two different bar codes may indicate that the two genetic variations are derived from two separate strands of DNA, reflecting a trans relationship.
  • the detection of two different genetic variations tagged with the same bar codes may indicate that the two genetic variations are from the same strand of DNA, reflecting a cis relationship.
  • Phase information may be important for the characterization of a polynucleotide fragment, particularly if the polynucleotide fragment is derived from a subject at risk of, having, or suspected of a having a particular disease or disorder (e.g., hereditary recessive disease such as cystic fibrosis, cancer, etc.).
  • the information may be able to distinguish between the following possibilities: (1) two genetic variations within the same gene on the same strand of DNA and (2) two genetic variations within the same gene but located on separate strands of DNA.
  • Possibility (1) may indicate that one copy of the gene is normal and the individual is free of the disease, while possibility (2) may indicate that the individual has or will develop the disease, particularly if the two genetic variations are damaging to the function of the gene when present within the same gene copy.
  • the phasing information may also be able to distinguish between the following possibilities: (1) two genetic variations, each within a different gene on the same strand of DNA and (2) two genetic variations, each within a different gene but located on separate strands of DNA.
  • Methods provided herein may also be used to prepare polynucleotides contained within cells in a manner that enables cell-specific information to be obtained.
  • the methods enable detection of genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) from very small samples, such as from samples comprising about 10-100 cells.
  • about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • at least about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • at most about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
  • a method comprises partitioning a cellular sample (or crude cell extract) such that at most one cell (or extract of one cell) is present per partition, lysing the cells, fragmenting the polynucleotides contained within the cells by any of the methods described herein, attaching the fragmented polynucleotides to barcodes, pooling, and sequencing.
  • the barcodes and other reagents may be contained within a partition (e.g., a capsule). These capsules may be loaded into another partition (e.g., a well) before, after, or concurrently with the loading of the cell, such that each cell is contacted with a different capsule.
  • This technique may be used to attach a unique barcode to polynucleotides obtained from each cell.
  • the resulting tagged polynucleotides may then be pooled and sequenced, and the barcodes may be used to trace the origin of the polynucleotides. For example, polynucleotides with identical barcodes may be determined to originate from the same cell, while polynucleotides with different barcodes may be determined to originate from different cells.
  • the methods described herein may be used to detect the distribution of oncogenic mutations across a population of cancerous tumor cells.
  • some tumor cells may have a mutation, or amplification, of an oncogene (e.g., HER2, BRAF, EGFR, KRAS) in both alleles (homozygous), others may have a mutation in one allele (heterozygous), and still others may have no mutation (wild-type).
  • an oncogene e.g., HER2, BRAF, EGFR, KRAS
  • the methods described herein may be used to detect these differences, and also to quantify the relative numbers of homozygous, heterozygous, and wild-type cells. Such information may be used, for example, to stage a particular cancer and/or to monitor the progression of the cancer and its treatment over time.
  • this disclosure provides methods of identifying mutations in two different oncogenes (e.g., KRAS and EGFR). If the same cell comprises genes with both mutations, this may indicate a more aggressive form of cancer. In contrast, if the mutations are located in two different cells, this may indicate that the cancer is more benign, or less advanced.
  • oncogenes e.g., KRAS and EGFR
  • a sample may comprise a cell, mRNA, or cDNA reverse transcribed from mRNA.
  • the sample may be a pooled sample, comprising extracts from several different cells or tissues, or a sample comprising extracts from a single cell or tissue.
  • Cells may be placed directly into a partition (e.g., a microwell) and lysed. After lysis, the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing cDNA may provide an indication of the abundance of a particular transcript in a particular cell over time, or after exposure to a particular condition.
  • a partition e.g., a microwell
  • the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing
  • the methods presented above provide several advantages over current polynucleotide processing methods.
  • Second, the methods may be carried out in microfluidic devices, which have a low cost and can be easily fabricated.
  • Third, the controlled fragmentation of the target polynucleotides allows the user to produce polynucleotide fragments with a defined and appropriate length. This aids in partitioning the polynucleotides and also reduces the amount of sequence information loss due to the present of overly-large fragments.
  • the methods and systems also provide a facile workflow that maintains the integrity of the processed polynucleotide.
  • the use of restriction enzymes enables the user to create DNA overhangs (“sticky ends”) that may be designed for compatibility with adapters and/or barcodes.
  • compositions, systems, devices, and kits provided in this disclosure may be used to partition polynucleotides, including whole chromosomes, from cells.
  • a single cell or a plurality of cells e.g., 2, 10, 50, 100, 1000, 10000, 25000, 50000, 100000, 500000, 1000000, or more cells
  • lysis buffer and proteinase K e.g., 1, 10, 50, 100, 1000, 10000, 25000, 50000, 100000, 500000, 1000000, or more cells
  • Utilization of a plurality of cells will enable polynucleotide phasing, for example, by partitioning each polynucleotide to be analyzed in its own partition.
  • the cell lysate is partitioned, for example by flow focusing the cell lysate into a capsule. If phasing is to be performed, flow focusing is performed such that each capsule comprises only a single analyte (e.g., a single chromosome), or only a single copy of any particular chromosome (e.g., one copy of a first chromosome and one copy of a second chromosome). In some cases, a plurality of chromosomes may be encapsulated within the same capsule, so long as the chromosomes are not the same chromosome. The encapsulation is performed under gentle flow, to minimize shearing of the polynucleotides.
  • flow focusing is performed such that each capsule comprises only a single analyte (e.g., a single chromosome), or only a single copy of any particular chromosome (e.g., one copy of a first chromosome and one copy of a second chro
  • the capsule may be porous, to allow washing of the contents of the capsule, and introduction of reagents into the capsule, while maintaining the polynucleotides (e.g., chromosomes) within the capsules.
  • the encapsulated polynucleotides e.g., chromosomes
  • the capsule shells protect the encapsulated polynucleotides (e.g., chromosomes) from shearing and further degradation. Of course, this method can also be applied to any other cellular component.
  • the capsule shell may be used to protect a polynucleotide from shearing.
  • a capsule may also be used as a partition to enable compartmentalized shearing of a polynucleotide or other analyte.
  • a polynucleotide may be encapsulated within a capsule and then subject to ultrasonic shear, or any other suitable shearing.
  • the capsule shell may be configured to remain intact under the shear, while the encapsulated polynucleotide may be sheared, but will remain within the capsule.
  • a hydrogel droplet may be used to accomplish the same end.
  • Barcoding methods via amplification-based barcoding schemes in partitions described herein may be useful generating barcode libraries from degraded samples such as, for example, fixed formalin-fixed, paraffin-embedded (FFPE) tissue sections.
  • Methods described herein may be capable of identifying that all amplicons within a partition originated from the same initial molecule. Indeed, with partition barcoding, information can be retained about a unique starting polynucleotide. Such identification may aid in determinations of library complexity as amplicons from different original molecules can be distinguished.
  • methods described herein can permit assessing unique coverage which may aid in determining variant calling sensitivity. These advantages may be particularly useful in cancer mutation detection and forensics.
  • Barcoding methods described herein may be useful in low polynucleotide input applications, such as, for example the sequencing of nucleic acids of circulating tumor cells (CTCs).
  • CTCs circulating tumor cells
  • MALBAC methods described herein within a partition may aid in obtaining good data quality in low polynucleotide input applications and/or aid in filtering out amplification errors.
  • kits comprising reagents for the generation of partitions.
  • the kit may comprise any suitable reagents and instructions for the generation of partitions and partitions within partitions.
  • a kit comprises reagents for generating capsules within droplets in an emulsion.
  • a kit may comprise reagents for generating capsules, reagents for generating an emulsion, and instructions for introducing the capsules into the droplets of the emulsion.
  • any suitable species may be incorporated into the droplets and/or into the capsule.
  • a kit of this disclosure may also provide any of these species, such as a polynucleotide comprising a barcode that is pre-partitioned.
  • the capsule may be designed to release its contents into the droplets of the emulsion upon the application of a stimulus.
  • a kit comprises reagents for generating capsules within capsules.
  • a kit may comprise reagents for generating inner capsules, reagents for generating outer capsules, and instructions for generating capsules within capsules.
  • any suitable species may be incorporated into the inner and/or outer capsules.
  • a kit of this disclosure may also provide any of these species, such as a polynucleotide comprising a barcode that is pre-partitioned.
  • the inner capsule may be designed to release its contents into the outer capsule upon the application of a stimulus.
  • this disclosure provides devices comprising partitions for the processing of analytes.
  • a device may be a microwell array, or a microspot array, as described elsewhere in this disclosure.
  • a device may formed in a manner that it comprises any suitable partition.
  • a device comprises a plurality of wells, or a plurality of spots.
  • any partition in a device may also hold other partitions, such as a capsule, a droplet in an emulsion, and the like.
  • a device may be formed from any suitable material.
  • a device is formed from a material selected from the group consisting of fused silica, soda lime glass, borosilicate glass, poly(methyl methacrylate), sapphire, silicon, germanium, cyclic olefin copolymer, polyethylene, polypropylene, polyacrylate, polycarbonate, plastic, and combinations thereof.
  • a device comprises channels for the flow of fluids into and between partitions. Any suitable channels may be used.
  • a device may comprise a fluid inlet and a fluid outlet. The inlet and outlet may be attached to liquid handling devices to introduce species into the device. The device may be sealed, before or after introduction of any species.
  • a device of this disclosure comprises a partition with an interior surface comprising a hydrophilic material.
  • a surface exterior to the partitions comprises a hydrophobic material.
  • a fluid flow path is coated with a hydrophobic or hydrophilic material.
  • compositions, libraries, methods, devices, and kits described herein for a particular use or purpose, including the various applications, uses, and purposes described herein.
  • the disclosure provides for the use of the compositions, methods, libraries, devices, and kits described herein in partitioning species, in partitioning oligonucleotides, in stimulus-selective release of species from partitions, in performing reactions (e.g., ligation and amplification reactions) in partitions, in performing nucleic acid synthesis reactions, in barcoding nucleic acid, in preparing polynucleotides for sequencing, in sequencing polynucleotides, in polynucleotide phasing, in sequencing polynucleotides from small numbers of cells, in analyzing gene expression, in partitioning polynucleotides from cells, in mutation detection, in neurologic disorder diagnostics, in diabetes diagnostics, in fetal aneuploidy diagnostics, in cancer mutation detection and forensics, in
  • This example provides methods for the manufacture of forked adapters comprising barcode sequences compatible with next generation sequencing technologies (e.g., ILLUMINA).
  • the barcode is placed in position 207 as depicted in FIG. 2 .
  • a single-stranded adapter-barcode polynucleotide sequence 401 comprising a first immobilization region 402 , a barcode region 403 , and a first sequencing primer region 404 is synthesized.
  • the barcode region 403 is a seven nucleotide random sequence synthesized by including equimolar concentrations of A, G, T, and C in each coupling step.
  • the single-stranded adapter-barcode polynucleotide 401 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 0.1 polynucleotides.
  • the droplets also comprise reagents for amplification of the single-stranded adapter-barcode polynucleotide 401 , by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts) and a DNA intercalating dye (e.g., ethidium bromide).
  • the reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification.
  • the polynucleotides are amplified and the reaction proceeds through an exponential phase of amplification 410 , which produces double-stranded products 405 , and a linear phase amplification 411 , which produces single-stranded products 406 .
  • the droplets are sorted on a fluorescence assisted cell sorter (FACS) 412 to collect droplets comprising amplified polynucleotides.
  • FACS fluorescence assisted cell sorter
  • a partially complementary universal sequence 407 is added to the partitions to generate a partially annealed fork structure 413 .
  • Partially complementary universal sequence 407 comprises a second immobilization region 408 and a second sequencing primer region 409 , the latter of which comprises a T overhang that is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
  • a single-stranded adapter-barcode polynucleotide sequence (e.g., FIG. 4 : 401 ) comprising a first immobilization region 402 , a barcode region 403 , and a first sequencing primer region 404 is synthesized, partitioned, amplified, and sorted as described in Example 1, or by any other method described in this disclosure.
  • Interfacial polymerization is performed on the droplet comprising the single-stranded adapter-barcode polynucleotide sequence, to generate a plurality of capsules comprising a library of single-stranded adapter-barcode polynucleotide sequences 406 , where each (or most) sequences in the library differ in the sequence of their respective barcode regions 403 .
  • a library of encapsulated single-stranded adapter-barcode polynucleotides is generated.
  • Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded), a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE), and a partially complementary universal sequence (e.g., FIG. 4 : 407 ).
  • a second mixture Z2 comprises the library of encapsulated single-stranded adapter-barcode polynucleotides, generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme.
  • Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
  • FIG. 5 illustrates a capsule within a capsule produced according to the method described above.
  • the outer capsule 501 comprises an inner capsule 502 and medium 504 .
  • the inner capsule 502 is one member of the library of encapsulated single-stranded adapter-barcode polynucleotides.
  • inner capsule 502 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 503 , which can be used to attach the same barcode to a polynucleotide within a partition, such as outer capsule 501 .
  • the medium 504 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 504 comprises target polynucleotide 505 , the partially complementary universal sequence 506 , and the enzyme mix 507 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
  • the enzymes Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture inner capsule 502 , releasing its contents into outer capsule 501 . The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide.
  • the single-stranded adapter-barcode polynucleotide 503 hybridizes with the partially complementary universal sequence 506 , forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide.
  • the thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide.
  • the outer capsule 501 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced. Additional preparation steps (e.g., bulk amplification, size selection, etc.) may then be performed as needed prior to sequencing.
  • mixture Z1 comprises multiple versions of the partially complementary universal sequence 506 , where each version has its own sample-specific barcode.
  • PCR can also be used to accomplish this step, as described elsewhere in this disclosure.
  • a library of encapsulated single-stranded adapter-barcode polynucleotides is generated as described in Example 2, or by any other suitable method described in this disclosure.
  • Target polynucleotides i.e., polynucleotides to be fragmented
  • the capsules comprising the target polynucleotides are configured to withstand ultrasonic stress.
  • the capsules comprising the target polynucleotides are exposed to ultrasonic stress (e.g., COVARIS Focused-Ultrasonicator) and the target polynucleotide is fragmented, generating fragmented target polynucleotide capsules.
  • ultrasonic stress e.g., COVARIS Focused-Ultrasonicator
  • a mixture Z1 is prepared, comprising the library of encapsulated single-stranded adapter-barcode polynucleotides (e.g., FIG. 4 : 406 ), the fragmented target polynucleotide capsules, a partially complementary universal sequence (e.g., FIG. 4 : 407 ), an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase), and appropriate buffers.
  • a capsule within capsule is generated according to the method described elsewhere in this disclosure, such as flow focusing.
  • FIG. 6 illustrates capsules within a capsule produced according to the methods described above.
  • the outer capsule 601 comprises a plurality of inner capsules 602 and 605 and medium 604 .
  • the inner capsules 602 and 605 include capsules comprising single-stranded adapter-barcode polynucleotides 603 and capsules comprising fragmented target polynucleotide 606 , respectively.
  • Inner capsule 602 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 603 , which can be used to attach the same barcode to a polynucleotide within a partition, such as the fragmented polynucleotides 606 contained within inner capsules 605 .
  • the medium 604 contains the contents of mixture Z1, described above. More specifically, medium 604 comprises a partially complementary universal sequence 607 , an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 608 , and appropriate buffers.
  • an enzyme mixture T4 polymerase, Taq polymerase, and a thermostable ligase
  • Inner capsules 605 comprising fragmented target polynucleotides 606 are exposed to a stimulus to rupture them and release their contents into the contents of outer capsule 601 .
  • the T4 polymerase blunts the ends of the fragmented target polynucleotides; the Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide.
  • the T4 polymerase and Taq polymerase is then heat-inactivated and a stimulus is applied to release the contents of inner capsule 602 into outer capsule 601 .
  • the single-stranded adapter-barcode polynucleotide 603 hybridizes with the partially complementary universal sequence 607 , forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide.
  • the thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating a barcoded target polynucleotide.
  • the outer capsule 601 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
  • Z1 can comprise multiple versions of the partially complementary universal sequence 607 .
  • this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
  • FIG. 7 provides an example of a product (or intermediate) that may be generated according to the methods of this example.
  • a hairpin adapter 701 (SEQ ID NO: 2) is shown that can be used as a precursor to a forked-adapter as described elsewhere in this disclosure.
  • the hairpin adapter is synthesized as a single-stranded amplification product utilizing SPIA.
  • the hairpin adapter 701 comprises a double-stranded region 702 , a 3′-T overhang for AT ligation 703 , and a region that can be cut by a restriction enzyme 704 (i.e., between positions 33 and 34).
  • the hairpin adapter may comprise a barcode region and functional regions, such as immobilization regions and regions for annealing of sequencing primers.
  • FIG. 8 a SEQ ID NOs: 3-4.
  • the adapter is cut by introducing an oligonucleotide sequence complementary to the region to be cut and exposing the annealed adapter to a restriction enzyme. Ligation of the forked-adapter region depicted in FIG. 8 a to a target polynucleotide results in the structure depicted in FIG. 8 b (SEQ ID NOs: 5-6).
  • FIG. 8 the underlined portions of the sequences in FIG.
  • SEQ ID NOs: 5-6 are then amplified by polymerase chain reaction to produce SEQ ID NO: 7 (amplification product of SEQ ID NO: 5) and SEQ ID NO: 8 (amplification product of SEQ ID NO: 6), shown in FIG. 8 c .
  • SEQ ID NO: 7 represents an amplification product of SEQ ID NO: 5 that adds a first immobilization sequence (underlined 5′ portion) and a second immobilization sequence (underlined 3′ portion) to SEQ ID NO: 5.
  • SEQ ID NO: 8 represents an amplification product of SEQ ID NO: 6 that replaces the unhybridized portions of SEQ ID NO: 6 with different sequences (underlined 3′ portion and underlined 5′ portion). Additionally, SEQ ID NO: 8 includes a six nucleotide barcode (TAGTGC; bolded) within the 5′ unhybridized region of the polynucleotide.
  • the amplification product therefore comprises barcoded target polynucleotide sequence (represented by 111), immobilization sequences, and a barcode.
  • FIG. 9 a shows the forked-adapter in single-stranded format (SEQ ID NO: 11), where the single stranded format is capable of forming a hairpin structure. Cutting the hairpin structure at the position designated by the asterisk yields the forked adapter shown in FIG. 9 a.
  • FIG. 9 c The template for the SPIA will be the sequence shown in FIG. 9 c (SEQ ID NO: 12).
  • R represents a region of RNA.
  • FIG. 9 d shows the hairpin structure formed by the sequence in FIG. 9 c .
  • the sequence in FIG. 9 d (SEQ ID NO: 12) is treated with polymerase to add nucleotides to the 3′ end, generating the sequence shown in FIG. 9 e (SEQ ID NO: 13).
  • the sequence in FIG. 9 e (SEQ ID NO: 13) is then treated with RNase H, which degrades RNA hybridized to DNA, yielding the sequence in FIG. 9 f (SEQ ID NO: 14).
  • Strand displacement SPIA is then performed on SEQ ID NO: 14.
  • the primer in the strand displacement amplification is of the form RRRRRRRRRRRRR (i.e., R 13 ).
  • This primer is an RNA primer that is one base longer than the unhybridized 3′ terminus of SEQ ID NO: 14 (i.e., N 12 ) ( FIG. 9 f ). More specifically, as shown in FIG. 9 f , the 3′ terminus of SEQ ID NO: 14 contains twelve N nucleotides.
  • the RNA primer contains 13 nucleotides. Nucleotides 2-13 of the RNA primer are complementary with the twelve unhybridized N nucleotides of SEQ ID NO: 14.
  • Nucleotide 1 of the RNA primer is complementary with the first hybridized base (going from 3′ to 5′), in this case, T.
  • the RNA primer displaces the A and generates the double-stranded extension product shown in FIG. 9 g (SEQ ID NOs: 15-16). Because only one primer is present, the reaction produces multiple copies of the single-stranded product.
  • the single-stranded amplification products are treated with RNase H to generate the single-stranded amplification products shown in FIG. 9 h (SEQ ID NO: 17).
  • FIG. 9 i shows this sequence in 5′-3′ format (SEQ ID NO: 17).
  • FIG. 9 j shows this sequence in hairpin format (SEQ ID NO 17).
  • the hairpin adapter shown in FIG. 9 j is then ligated to a fragmented polynucleotide with a 3′-A overhang.
  • the hairpin is cleaved between the A and C residues separated by the curved line in FIG. 9 j by adding an oligonucleotide complementary to that region and cutting with a restriction enzyme. This generates a forked adapter.
  • PCR amplification is then conducted, as described in Example 4, to add immobilization regions and barcodes to the forked adapter that is attached to the target polynucleotide.
  • FIG. 10 a shows the exemplary forked adapter provided in FIG. 8 a .
  • this adapter may be ligated to a target polynucleotide and then an amplification reaction may be performed to add additional functional sequences, including a barcode.
  • a barcode (and other functional sequences) may also be incorporated directly into the forked adapter, prior to attachment of the forked adapter to the target polynucleotide.
  • FIG. 10 b shows the forked adapter of FIG. 10 a , with the addition of a first immobilization region (underlined) and a seven nucleotide barcode region (bold/underline; “N”).
  • the barcoded forked adapter of FIG. 10 b is produced by first synthesizing SEQ ID NO: 18 as a single strand.
  • the diversity in the barcode region is generated using an equimolar mixture of A, G, T, and C, as described throughout this disclosure.
  • Droplet-based PCR is performed, as described in Example 1. However, one DNA primer and one RNA primer are used to amplify SEQ ID NO: 18 in the droplets.
  • the amplification is conducted in the presence of an intercalating dye, and droplets comprising amplified SEQ ID NO: 18 are isolated, as described in Example 1.
  • FIG. 10 c shows the double-stranded amplification product.
  • the underlined portion of SEQ ID NO: 19 is an RNA strand derived from the RNA primer.
  • sequences shown in FIG. 10 c are then treated with RNase H, which digests the underlined RNA region, yielding the construct shown in FIG. 10 d .
  • RNase H which digests the underlined RNA region
  • a partially complementary universal sequence (SEQ ID NO: 21) is added to the construct shown in FIG. 10 d , producing the product shown in FIG. 10 e .
  • SEQ ID NO: 21 partially complementary universal sequence
  • a dual-index read is a read of both strands of a double-stranded fragment, using barcodes attached to each strand.
  • FIG. 11 shows an example of the synthesis of barcodes for a dual-indexing approach and an example use of the barcodes in a capsules in a capsule configuration.
  • a first single-stranded adapter-barcode polynucleotide sequence 1101 comprising a first immobilization region 1102 , a first barcode region 1103 , and a first sequencing primer region 1104 is synthesized.
  • a second single-stranded adapter-barcode polynucleotide sequence 1131 comprising a second immobilization region 1132 , a second barcode region 1133 , and a second sequencing primer region 1134 is synthesized.
  • barcode regions 1103 and 1133 are of the same sequence. In other cases, barcode regions 1103 and 1133 are of different sequences or of partially different sequences.
  • the single-stranded adapter-barcode polynucleotides 1101 ( FIG. 11 a ) and 1131 ( FIG. 11 b ) are, in parallel, diluted into aqueous droplets in a water-in-oil emulsion.
  • the droplets also comprise reagents for amplification of the single-stranded adapter-barcodes polynucleotide 1101 ( FIG. 11 a ) and 1131 ( FIG. 11 b ) respectively, by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts) and a DNA intercalating dye (e.g., ethidium bromide).
  • asymmetric PCR e.g., polymerase, primers, dNTPs, buffer, salts
  • a DNA intercalating dye e.g., ethidium bromide
  • the reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification.
  • the polynucleotides 1101 ( FIG. 11 a ) and 1131 ( FIG. 11 b ) are amplified and the reaction proceeds through an exponential phase of amplification 1110 , which produces double-stranded products 1105 ( FIG. 11 a ) and 1135 ( FIG. 11 b ), and a linear phase amplification 1111 , which produces single-stranded products 1106 ( FIG. 11 a ) and 1136 ( FIG. 11 b ) respectively.
  • the droplets are sorted on a fluorescence assisted cell sorter (FACS) 1112 to collect droplets comprising amplified polynucleotides.
  • FACS fluorescence assisted cell sorter
  • Interfacial polymerization is then performed on the droplets comprising the single-stranded adapter-barcode polynucleotide sequences 1106 and 1136 droplets respectively, to generate two types of capsules 1120 ( FIG. 11 a ) and 1150 ( FIG. 11 b ), each comprising one of single-stranded adapter barcode polynucleotide sequences 1106 or 1136 respectively.
  • Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded) 1170 and a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE).
  • a second mixture Z2 comprises capsules 1120 and 1180 , generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme.
  • Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
  • FIG. 11 c illustrates capsules within a capsule produced according to the method described above.
  • the outer capsule 1160 comprises capsules 1120 and 1150 and medium 1190 .
  • capsules 1120 and 1150 each comprise multiple copies of single-stranded adapter-barcode polynucleotides 1106 and 1136 , respectively, and can be used to attach barcodes 1103 and 1133 to a polynucleotide within a partition, such as target polynucleotide 1170 in medium 1190 of outer capsule 1160 .
  • the medium 1190 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 1190 comprises target polynucleotide 1170 and the enzyme mix 1180 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
  • the enzymes Upon generation of the capsules within a capsule, and exposure of the capsules within the capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture capsules 1120 and 1150 , releasing their contents into medium 1190 of outer capsule 1160 . The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide.
  • the single-stranded adapter-barcode polynucleotide 1106 hybridizes with single-stranded adapter-barcode polynucleotide 1136 , forming a forked adapter, comprising barcode regions 1103 and 1133 , with a 3′-T overhang that is compatible with the 3′-A overhang (not shown) on the fragmented target polynucleotide.
  • the thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide.
  • the outer capsule 1160 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
  • PCR can also be used to accomplish this step, as described elsewhere in this disclosure.
  • a single-stranded adapter-barcode sequence 1401 comprising a first immobilization region 1402 , a barcode region 1403 , and a first sequencing primer region 1404 is synthesized.
  • the single-stranded adapter-barcode sequence 1401 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 1 polynucleotide.
  • the droplets also comprise first beads 1405 that are linked, via a photolabile linker, to one or more copies of an RNA primer 1406 complementary to a sequence comprised in the first sequencing primer region 1404 ; a DNA primer complementary to a sequence (not shown) comprised in the first immobilization region 1402 ; and reagents necessary for amplification (e.g., polymerase, dNTPs, buffer, salts) of single-stranded adapter-barcode sequence 1401 .
  • the polynucleotides are amplified 1407 which produces double-stranded products 1408 both attached to the first beads 1405 to form structure 1420 and in solution (not shown).
  • the emulsion is then broken and the emulsion components are pooled to form a product mixture.
  • the liberated beads are then washed 1409 (via centrifugation) several times with appropriate medium, treated with sodium hydroxide (NaOH) 1410 to denature the double-stranded products attached to the first beads 1405 , and then further washed 1411 .
  • the resulting structure 1430 comprises a single-stranded complement 1412 to the single-stranded adapter-barcode sequence 1401 , comprising a complementary immobilization region 1413 , a complementary barcode region 1414 , and a complementary sequencing primer region 1415 .
  • the complementary sequencing primer region 1415 comprises the RNA primer 1406 .
  • Structure 1430 is then resuspended in an appropriate medium.
  • second beads 1416 that comprise one or more copies of a DNA polynucleotide 1417 complementary to the complementary immobilization region 1413 are then added to the medium. Via the complementary DNA polynucleotide 1417 and the complementary immobilization region 1413 of the single-stranded complement 1412 , the second beads 1416 bind to the single-stranded complement 1412 .
  • the single stranded complement is now bound at one end to first bead 1405 and at its other end second bead 1416 to form structure 1440 .
  • structure 1440 is then centrifuged 1418 using a glycerol gradient to separate structure 1440 from structure 1430 not comprised in structure 1440 .
  • the second beads 1416 are magnetic
  • a magnetic separation may be used as an alternative.
  • the product is then treated with NaOH 1419 to denature the single-stranded complement 1412 from the second bead 1416 , resulting in regeneration of structure 1430 .
  • Structure 1430 is then subject to several rounds of washing (via centrifugation) to remove second beads 1416 .
  • Single-stranded complement 1412 attached to structure 1430 , represents a single-stranded barcode adapter.
  • single-stranded complement 1412 can be used to generate a forked adapter.
  • the single-stranded complement 1412 is then released 1424 from structure 1430 with light and then combined 1425 with a universal complementary sequence 1426 or is first combined 1425 with a universal complementary sequence 1426 and then released 1424 from structure 1430 .
  • RNAase H is used to digest the RNA primer 1406 of the single-stranded complement 1412 and a Type IIs restriction enzyme is used to generate a single base T overhang on the universal complementary sequence 1426 .
  • the T overhang is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
  • a single-stranded adapter-barcode sequence 1501 comprising a first immobilization region 1502 , a barcode region 1503 , and a first sequencing primer region 1504 is synthesized.
  • the single-stranded adapter-barcode sequence 1501 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 1 polynucleotides.
  • the droplets also comprise first beads 1505 that are linked, via a photolabile linker, to one or more copies of an RNA primer 1506 complementary to a sequence comprised in the first immobilization region 1502 ; a DNA primer complementary to a sequence (not shown) comprised in the first sequencing primer region 1502 ; and reagents necessary for amplification (e.g., polymerase, dNTPs, buffer, salts) of single-stranded adapter-barcode sequence 1501 .
  • the polynucleotides are amplified 1507 which produces double-stranded products 1508 both attached to the first beads 1505 to form structure 1520 and in solution (not shown).
  • the emulsion is then broken and the emulsion components are pooled to form a product mixture.
  • the liberated beads are then washed 1509 (via centrifugation) several times with appropriate medium, treated with sodium hydroxide (NaOH) 1510 to denature the double-stranded products attached to the first beads 1505 , and then further washed 1511 .
  • the resulting structure 1430 comprises a single-stranded complement 1512 to the single-stranded adapter-barcode sequence 1501 , comprising a complementary immobilization region 1513 , a complementary barcode region 1514 , and a complementary sequencing primer region 1515 .
  • the complementary sequencing primer region 1515 comprises the RNA primer 1506 .
  • Structure 1530 is then resuspended in an appropriate medium.
  • second beads 1516 that comprise one or more copies of a DNA polynucleotide 1517 complementary to the complementary sequencing primer region 1515 are then added to the medium.
  • the second beads 1416 bind to the single-stranded complement 1512 .
  • the single stranded complement is now bound at one end to first bead 1505 and at its other end second bead 1516 to form structure 1540 .
  • structure 1540 is then centrifuged 1518 using a glycerol gradient to separate structure 1540 from structure 1530 not comprised in structure 1540 .
  • the second beads 1516 are magnetic
  • a magnetic separation may be used as an alternative.
  • the product is then treated with NaOH 1519 to denature the single-stranded complement 1512 from the second bead 1516 , resulting in regeneration of structure 1530 .
  • Structure 1530 is then subject to several rounds of washing (via centrifugation) to remove second beads 1516 .
  • Single-stranded complement 1512 attached to structure 1530 , represents a single-stranded barcode adapter.
  • single-stranded complement 1512 can be used to generate a forked adapter.
  • the single-stranded complement 1512 is then optionally released 1524 from structure 1530 with light and then combined 1525 with a universal complementary sequence 1526 .
  • a Type IIs restriction enzyme is used to generate a single base T overhang on the universal complementary sequence 1526 .
  • the T overhang is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
  • structure 1600 comprising a magnetic bead ( 1601 )-bound single-stranded adapter-barcode sequence 1602 is produced according to methods described in Example 8, Example 9, or any other method described herein.
  • structure 1600 is partitioned into capsules (or alternatively, another emulsion) 1620 by methods described herein, for example, interfacial polymerization.
  • the capsules 1620 also comprise reagents for amplification of the single-stranded adapter-barcode sequence 1602 , by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts).
  • the reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification.
  • Single-stranded adapter-barcode sequence 1602 is amplified 1603 and the reaction proceeds through a linear phase amplification 1604 , which produces single-stranded adapter product 1605 , complementary to single-stranded barcode adapter-template 1602 .
  • capsules 1620 comprise both single-stranded adapter 1605 in solution and magnetic bead ( 1601 )-bound single-stranded adapter-barcode sequence 1602 .
  • Capsules 1620 are then separated from those not comprising beads (and thus templates 1602 and single-stranded adapters 1605 ) by magnetic separation 1606 .
  • Capsules 1620 may be ruptured and forked adapters generated as described in Example 9.
  • structure 1700 comprising a magnetic bead ( 1701 )-bound single-stranded adapter-barcode sequence 1702 is produced according to methods described in Example 8, Example 9, or any other method described herein.
  • Interfacial polymerization is performed on the droplet comprising the structure 1700 , to generate a capsule 1704 comprising single-stranded adapter-barcode sequence 1702 attached, via a photolabile linker, to a bead 1701 .
  • Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded), a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE), and a partially complementary universal sequence.
  • a second mixture Z2 comprises capsule 1704 generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme.
  • Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
  • FIG. 17 illustrates a capsule within a capsule produced according to the method described above.
  • the outer capsule 1703 comprises an inner capsule 1704 and medium 1705 .
  • the inner capsule 1704 is one member of a library of encapsulated, bead-bound single-stranded barcode adapters.
  • inner capsule 1704 comprises multiple copies of structure 1700 , which can be used to generate a free single-stranded adapter-barcode sequence 1702 and attach the same barcode adapter to a polynucleotide within a partition, such as outer capsule 1703 .
  • the medium 1705 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 1705 comprises target polynucleotide 1706 , the partially complementary universal sequence 1707 , and the enzyme mix 1708 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
  • the enzymes Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture inner capsule 1704 , releasing its contents into outer capsule 1703 . The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide.
  • the single-stranded adapter-barcode sequence 1702 hybridizes with the partially complementary universal sequence 1707 and is released from the bead with light, forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide.
  • the thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide.
  • the outer capsule 1703 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced. Additional preparation steps (e.g., bulk amplification, size selection, etc.) may be performed as needed prior to sequencing.
  • Z1 can comprise multiple versions of the partially complementary universal sequence 1707 .
  • this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
  • structure 1800 comprising a magnetic bead ( 1801 )-bound single-stranded adapter-barcode sequence 1802 is produced according to methods described in Example 8, Example 9, or any other method described herein.
  • Interfacial polymerization is performed on the droplet comprising the structure 1800 , to generate a capsule 1803 comprising single-stranded adapter-barcode sequence 1802 attached, via a photolabile linker, to a bead 1801 .
  • Target polynucleotides i.e., polynucleotides to be fragmented
  • the capsules 1804 comprising the target polynucleotides are configured to withstand ultrasonic stress.
  • the capsules 1804 comprising the target polynucleotides are exposed to ultrasonic stress (e.g., COVARIS Focused-Ultrasonicator) and the target polynucleotide is fragmented, generating fragmented target polynucleotide capsules.
  • ultrasonic stress e.g., COVARIS Focused-Ultrasonicator
  • a mixture Z1 is prepared, comprising capsule 1803 , the fragmented target polynucleotide capsules 1804 , a partially complementary universal sequence 1805 , an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 1806 , and appropriate buffers.
  • a capsule within capsule is generated according to the method described elsewhere in this disclosure, such as flow focusing.
  • FIG. 18 illustrates capsules within a capsule produced according to the methods described above.
  • the outer capsule 1807 comprises capsules 1803 and 1804 and medium 1808 .
  • the inner capsules 1803 and 1804 include capsules comprising structure 1800 and capsules comprising fragmented target polynucleotide 1809 , respectively.
  • Inner capsule 1803 comprises multiple copies of structure 1800 , which can be used to generate a free single-stranded barcode adapter 1802 and attach the same barcode adapter to a polynucleotide within a partition, such as the fragmented polynucleotides 1809 contained within inner capsules 1804 .
  • the medium 1808 contains the contents of mixture Z1, described above. More specifically, medium 1808 comprises a partially complementary universal sequence 1805 , an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 1806 , and appropriate buffers.
  • an enzyme mixture T4 polymerase, Taq polymerase, and a thermostable ligase
  • Inner capsules 1804 comprising fragmented target polynucleotides 1809 are exposed to a stimulus to rupture them and release their contents into the contents of outer capsule 1807 .
  • the T4 polymerase blunts the ends of the fragmented target polynucleotides; the Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide.
  • the T4 polymerase and Taq polymerase is then heat-inactivated and a stimulus is applied to release the contents of inner capsule 1803 into outer capsule 1807 .
  • the single-stranded adapter-barcode sequence 1802 hybridizes with the partially complementary universal sequence 1805 and the adapter is released from the bead with light forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide.
  • the thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating a barcoded target polynucleotide.
  • the outer capsule 1807 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
  • Z1 can comprise multiple versions of the partially complementary universal sequence 1807 .
  • this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
  • a primer comprising SEQ ID NO: 36 is prepared.
  • the primer comprises a barcode region (designated “Barcode”), a primer sequencing region (designated “PrimingSeq”), and a eight-nucleotide variable region (designated as “ ”) that may comprise any combination of A, T, C, or G.
  • the primer shown in FIG. 19 is combined with a target polynucleotide (indicated by the loop in FIG.
  • a polymerase e.g., Vent, exo+DeepVent, exo ⁇ DeepVent
  • a partition e.g., a capsule, droplet of an emulsion, etc.
  • a non strand-displacing polymerase e.g., Taq, PfuUltra
  • MALBAC cycling conditions are known and are, described for example, in Zong et al., Science, 338(6114), 1622-1626 (2012), which is incorporated herein by reference, in its entirety.
  • a looped MALBAC product is produced as shown in FIG. 19 b as SEQ ID NO: 23.
  • the looped MALBAC product comprises the original primer shown in FIG. 19 a , the target polynucleotide to be barcoded oriented in a loop, and a region complementary to and hybridized to the original primer sequence.
  • the partition is broken and the contents recovered. In some cases, a plurality of partitions are generated. The partitions are collectively broken, the contents of each recovered, and then pooled.
  • the generated MALBAC product shown in FIG. 19 b is treated with a restriction enzyme (e.g., BfuC1 or similar) to generate a 4-basepair overhang (in this case, GATC shown in italics) on the MALBAC product.
  • a restriction enzyme e.g., BfuC1 or similar
  • SEQ ID NO: 24 SEQ ID NO: 24
  • SEQ ID NO: 37 SEQ ID NO: 37
  • the forked adapter shown in FIG. 19 d as SEQ ID NO: 25 and SEQ ID NO: 37, comprising an overhang (in this case, CTAG shown in bold) complementary to the overhang generated on the MALBAC product.
  • the forked adapter is mixed with the MALBAC product in FIG. 19 c and the complementary regions hybridize.
  • thermostable ligase is used to ligate the forked adapter and MALBAC product together to form the desired structure FIG. 19 e as SEQ ID NO: 26.
  • Additional amplification methods e.g., PCR
  • additional regions e.g., immobilization regions, additional barcodes, etc.
  • basepair overhangs e.g., 1 basepair overhang-10 basepair overhang
  • Restriction enzymes used to generate these overhangs may be used as an alternative, including those described herein, where desired.
  • a two basepair overhang is generated on the MALBAC product using Taq ⁇ I.
  • MALBAC product 1900 comprises an RNA primer sequence 1901 placed 5′ of a barcode region 1902 .
  • MALBAC product 1900 also comprises a sequencing primer region 1903 , the target polynucleotide 1904 , a complementary sequencing primer region 1905 , a complementary barcode region 1906 , and a region 1907 complementary to the RNA primer sequence 1901 .
  • MALBAC product 1900 is treated with an RNAse H 1908 and the RNA primer region sequence 1901 is digested to yield a 2-6 basepair overhang 1909 on MALBAC product 1900 to give structure 1920 .
  • a universal complementary region 1910 is then added to structure 1910 that comprises a region complementary to the overhang on structure 1910 .
  • Universal complementary region 1910 then hybridizes with structure 1920 and a thermostable ligase is used to ligate universal complementary region 1910 to structure 1920 .
  • a template 2000 comprising a barcode region is combined with agents 2001 necessary for PCR into a capsule 2002 , using, for example interfacial polymerization or any other method described herein.
  • PCR is used to generate a MALBAC primer from the template 2000 .
  • the capsule 2000 is encapsulated into an outer capsule 2003 that also comprises a mixture 2004 that comprises a target polynucleotide 2005 to be barcoded and reagents 2006 necessary for MALBAC amplification (e.g., DeepVent polymerase, dNTPs, buffer).
  • reagents 2006 necessary for MALBAC amplification e.g., DeepVent polymerase, dNTPs, buffer.
  • Capsule 2002 is broken upon proper exposure of capsule 2002 to a stimulus designed to rupture capsule 2002 , the contents of capsule 2002 mix with those of mixture 2004 .
  • MALBAC amplification of the target polynucleotide 2005 commences to produce a MALBAC product similar to that described as that shown as 1900
  • the outer capsule 2003 is then broken with the appropriate stimulus and the contents recovered.
  • the MALBAC product is then treated with an appropriate restriction enzyme and coupled to a forked adapter in a matter as described in Example 13. Additional downstream preparation steps (e.g., bulk amplification, size selection, etc.) are then performed as needed.
  • MALBAC primer 2100 is prepared.
  • MALBAC primer 2100 comprises a sequence priming region 2101 and an 8-nucleotide variable region 1902 .
  • Primer 2100 is combined with target polynucleotide 2103 , along with a polymerase (e.g., Vent, exo+DeepVent, exo ⁇ DeepVent) possessing of strand-displacement activity into a partition (e.g., a capsule, emulsion, etc.).
  • a non strand-displacing polymerase e.g., Taq, PfuUltra
  • the partition is then subject to MALBAC amplification 2104 .
  • a looped MALBAC product 2110 is produced and comprises sequencing priming region 2101 , target polynucleotide 2103 , and a complementary sequence priming region 2105 . Shown in linear form 2120 in FIG. 21 b , MALBAC product 2110 is then contacted with another primer 2130 that comprises a sequencing primer region 2106 , a barcode region 2107 , and an immobilization region 2108 .
  • Primer 2130 is produced using asymmetric digital PCR. Using a single cycle of PCR, the primer is used to generate double-stranded product 2140 that comprises primer 2130 , and, thus, barcode region 2107 .
  • Double-stranded product 2140 may be then denatured and subsequently contacted with another primer 2150 shown in FIG. 21 c .
  • Primer 2150 comprises a barcode region 2109 , a sequencing primer region 2111 , and an immobilization region 2112 .
  • additional rounds of PCR can add the barcode region 2109 to the end of the target polynucleotide that attached to barcode region 2107 .
  • Additional downstream preparation steps e.g., bulk amplification, size selection, etc. are then performed as needed.
  • a primer template 2200 comprising a barcode region is combined with agents 2201 necessary for PCR into a capsule 2202 , using, for example interfacial polymerization or any other method described herein. PCR is then used to generate a primer from template 2200 .
  • the capsule 2200 is encapsulated into an outer capsule 2003 that also comprises a mixture 2204 that comprises a target polynucleotide 2205 to be barcoded, reagents 2206 necessary for MALBAC amplification (e.g., DeepVent polymerase, dNTPs, buffer), and a MALBAC primer 2207 that does not comprise a barcode (similar to MALBAC primer 2100 described in Example 15).
  • MALBAC amplification of the target polynucleotide 2205 commences to produce a MALBAC product similar to that described as that shown as 2110 in FIG. 21 a .
  • Capsule 2202 is then broken upon proper exposure of capsule 2202 to a stimulus designed to rupture capsule 2202 , the contents of capsule 2202 mix with those of mixture 2004 .
  • a single cycle of PCR commences using the primer generated from template 2200 to generate a barcoded product, similar to that described in Example 15.
  • Outer capsule 2203 is then broken with the appropriate stimulus and the contents recovered. Additional downstream preparation steps (e.g., bulk amplification, size selection, addition of additional barcodes, etc.) are then performed as needed.
  • Additional downstream preparation steps e.g., bulk amplification, size selection, addition of additional barcodes, etc.
  • a single-stranded adapter-barcode polynucleotide sequence 2300 is synthesized, partitioned, amplified, and sorted as described in Example 1, or by any other method described in this disclosure. Interfacial polymerization is performed on the droplet comprising the single-stranded adapter-barcode polynucleotide sequence, to generate a capsule 2301 .
  • Mixture Z1 comprises a target polynucleotide 2302 (i.e., a polynucleotide to be fragmented and barcoded), a transposome 2303 , and a partially complementary universal sequence 2304 .
  • a second mixture Z2 comprises capsule 2301 , generated as described above and reagents 2305 necessary for PCR as described elsewhere herein.
  • FIG. 23 illustrates a capsule within a capsule produced according to the method described above.
  • the outer capsule 2306 comprises capsule 2301 and medium 2307 .
  • Capsule 2301 is one member of a library of encapsulated single-stranded adapter-barcode polynucleotides.
  • capsule 2301 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 2300 , which can be used to attach the same barcode to a polynucleotide within a partition, such as outer capsule 2306 .
  • the medium 2307 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 2307 comprises target polynucleotide 2302 , the partially complementary universal sequence 2304 , and the reagents 2305 necessary for PCR, including a hot start Taq.
  • the transposome Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the transposome process the target polynucleotide. More specifically, the transposase fragments the target polynucleotide via tagmentations and tags it with a common priming sequence. The tagged target polynucleotide is then heated to fill in any gap in the target nucleotide generated by the transposase. The transposase is then heat inactivated and a stimulus is used to rupture inner capsule 2301 , releasing its contents into outer capsule 2306 . The hot start Taq is activated by heating the outer capsule 2306 to 95° C.
  • the reaction proceeds with limited cycle PCR to add single-stranded adapter-barcode polynucleotide sequence 2300 to target polynucleotide 2302 .
  • the outer capsule 2306 is then ruptured and the target polynucleotides are sequenced.

Abstract

The present disclosure provides compositions, methods, systems, and devices for polynucleotide processing. Such polynucleotide processing may be useful for a variety of applications, including polynucleotide sequencing. In some cases, this disclosure provides methods for the generation of polynucleotide barcode libraries, and for the attachment of such polynucleotides to target polynucleotides.

Description

CROSS-REFERENCE
This application claims the benefit of U.S. Provisional Patent Application No. 61/762,435, filed Feb. 8, 2013, U.S. Provisional Patent Application No. 61/800,223, filed Mar. 15, 2013, U.S. Provisional Patent Application No. 61/840,403, filed Jun. 27, 2013, and U.S. Provisional Patent Application No. 61/844,804, filed Jul. 10, 2013, said applications are incorporated herein by reference in their entireties for all purposes.
The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Mar. 7, 2014, is named 43487706201SL.txt and is 14,795 bytes in size.
BACKGROUND
Polynucleotide barcodes have utility in numerous applications, including next generation sequencing techniques. Such barcodes generally contain unique identifier sequences, which can be extremely expensive to manufacture at sufficient diversity and scale. The cost of synthesizing a single polynucleotide barcode is a function of the cost per base during synthesis and the length of the polynucleotide. The cost of synthesizing a plurality of barcodes, each with a different sequence, is therefore equivalent to the cost per base, multiplied by the number of bases per molecule, multiplied by the number of molecules within the plurality of barcodes. Currently, it costs approximately $0.10 per base to synthesize a DNA sequence. For a barcode library of tens of thousands to millions of barcodes, this cost is prohibitive. Thus, there is a significant need for improved methods of generating libraries of barcodes.
SUMMARY
This disclosure provides methods, compositions, systems, and kits for the generation of polynucleotide barcodes and the use of such polynucleotide barcodes. Such polynucleotide barcodes may be used for any suitable application.
An aspect of the disclosure provides a library comprising one or more polynucleotides, each of the polynucleotides comprising a barcode sequence, wherein the polynucleotides are disposed within one or more partitions, and wherein the library comprises at least about 1,000 different barcode sequences.
In some cases, the barcode sequences are at least about 5 nucleotides in length. Also, the barcode sequences may be random polynucleotide sequences.
Moreover, the partitions may comprise, on average, about 1 polynucleotide, about 0.5 polynucleotides, or about 0.1 polynucleotides. The partitions may be droplets, capsules, wells or beads.
Furthermore, the library may comprise at least about 10,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 500,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 2,500,00 different barcode sequences, at least about 5,000,000 different barcode sequences, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
In some cases, the partitions may comprise multiple copies of the same polynucleotide.
Additionally, each of the polynucleotides may comprise a sequence selected from the group consisting of an immobilization sequence, an annealing sequence for a sequencing primer, and a sequence compatible for ligation with a target polynucleotide.
In some cases, each of the polynucleotides is a MALBAC primer.
Another aspect of the disclosure provides a method of synthesizing a library of polynucleotides comprising barcode sequences, the method comprising: a.) synthesizing a plurality of polynucleotides comprising barcode sequences; b.) separating the polynucleotides into a plurality of partitions, thereby generating partitioned polynucleotides; c.) amplifying the partitioned polynucleotides, thereby generating amplified polynucleotides; and d.) isolating partitions comprising amplified polynucleotides. In some cases, the synthesizing comprises including a mixture of adenine, thymine, guanine, and cytosine in a coupling reaction.
Moreover, the separating may comprise performing a limiting dilution, thereby generating diluted polynucleotides. In some cases, the separating further comprises partitioning said diluted polynucleotides.
Additionally, the amplifying may be performed by a method selected from the group consisting of polymerase chain reaction, asymmetric polymerase chain reaction, emulsion PCR (ePCR), ePCR including the use of a bead, ePCR including the use of a hydrogel, multiple annealing and looping-based amplification cycles (MALBAC), single primer isothermal amplification, and combinations thereof. In some cases, the amplifying is performed using an RNA primer and may include exposing the amplified polynucleotides to an RNAase H.
In some cases, each of said polynucleotides comprising barcode sequences is a MALBAC primer.
In some cases, the isolating may be performed by flow-assisted sorting.
Also, a hairpin structure may be formed from a polynucleotide selected from the group consisting of the polynucleotides comprising barcode sequences and the amplified polynucleotides. In some cases, a method may further comprise cutting the hairpin structure within an unannealed region.
Moreover, a polynucleotide selected from the group consisting of said polynucleotides comprising barcode sequences, said partitioned polynucleotides, and said amplified polynucleotides may be attached to a bead.
The method may further comprise annealing the amplified polynucleotides with a partially complementary sequence. The partially complementary sequence may comprise a barcode sequence.
The method may further comprise attaching at least one of the amplified polynucleotides to a target sequence. The target sequence may be fragmented. In some cases, the target sequence is fragmented by a method selected from the group consisting of mechanical shear and treatment with an enzyme. The mechanical shear may be induced by ultrasound. In some cases, the enzyme is selected from the group consisting of a restriction enzyme, a fragmentase, and a transposase. Additionally, the attaching may be performed by a method selected from the group consisting of ligation and amplification.
In some cases, the amplification is a MALBAC amplification performed with MALBAC primers, thereby generating a MALBAC amplification product. In some cases, the MALBAC primers comprise the amplified polynucleotides. In some cases, the MALBAC primers comprise polynucleotides that are not said amplified polynucleotides. In such cases, the method may further comprise attaching the MALBAC amplification product to the amplified polynucleotide.
Additionally, each of the partitions may comprise, on average, about 1 polynucleotide comprising a barcode sequence, 0.5 polynucleotides comprising barcode sequences, or 0.1 polynucleotides comprising barcode sequences. Moreover, the partitions may be selected from the group consisting of droplets, capsules, and wells.
In some cases, the library comprises at least about 1,000 different barcode sequences, at least about 10,000 different barcode sequences, at least about 100,000 different barcode sequences, at least about 500,000 different barcode sequences, at least about 1,000,000 different barcode sequences, at least about 2,500,00 different barcode sequences, at least about 5,000,000 different barcode sequences, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
In some cases, the partitions comprise multiple copies of the same polynucleotide comprising a barcode sequence.
Moreover, the polynucleotides comprising barcode sequences may comprise a sequence selected from the group consisting of an immobilization sequence, an annealing sequence for a sequencing primer, and a sequence compatible for ligation with a target polynucleotide.
An additional aspect of the disclosure provides a library comprising at least about 1,000 beads, wherein each bead of the at least about 1,000 beads comprises a different barcode sequence. In some cases, the different barcode sequence can be included in a polynucleotide comprising an immobilization sequence and/or an annealing sequence for a sequencing primer. In some cases, the different barcode sequence can be at least about 5 nucleotides or at least about 10 nucleotides in length. In some cases, the different barcode sequence can be a random polynucleotide sequence or can be generated combinatorially.
Moreover, each of the 1,000 beads can comprise multiple copies of the different barcode sequence. For example, each of the 1,000 beads may comprise at least about 100,000, at least about 1,000,000, or at least about 10,000,000 copies of the different barcode sequence. In some cases, the library can further comprise two or more beads comprising the same barcode sequence. In some cases, at least two beads of the 1,000 beads can comprise the same barcode sequence. Furthermore, the at least about 1,000 beads may comprise at least about 10,000 beads, or at least about 100,000 beads.
Also, the library can comprise at least about 1,000, at least about 10,000, at least about 100,000, at least about 1,000,000, at least about 2,500,000, at least about 5,000,000, at least about 10,000,000, at least about 25,000,000, at least about 50,000,000, or at least about 100,000,000 different barcode sequences.
In some cases, the at least about 1,000 beads can be distributed across a plurality of partitions. In some cases, the partitions can be droplets of an emulsion. In some cases, each bead of the 1,000 beads can be included in a different partition. In some cases, the different partition can be a droplet of an emulsion. In some cases, two or more beads of the 1,000 beads can be included in a different partition. In some cases, the different partition can be a droplet of an emulsion. In some cases, the 1,000 beads can be hydrogel beads.
An additional aspect of the disclosure provides for use of a library, composition, method, device, or kit described herein in partitioning species, in partitioning oligonucleotides, in stimulus-selective release of species from partitions, in performing reactions (e.g., ligation and amplification reactions) in partitions, in performing nucleic acid synthesis reactions, in barcoding nucleic acid, in preparing polynucleotides for sequencing, in sequencing polynucleotides, in mutation detection, in neurologic disorder diagnostics, in diabetes diagnostics, in fetal aneuploidy diagnostics, in cancer mutation detection and forenscics, in disease detection, in medical diagnostics, in low input nucleic acid applications, in circulating tumor cell (CTC) sequencing, in polynucleotide phasing, in sequencing polynucleotides from small numbers of cells, in analyzing gene expression, in partitioning polynucleotides from cells, or in a combination thereof.
INCORPORATION BY REFERENCE
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWINGS
The novel features of methods, compositions, systems, and devices of this disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of this disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the methods, compositions, systems, and devices of this disclosure are utilized, and the accompanying drawings of which:
FIG. 1 is schematically depicts an example forked adapter.
FIG. 2 schematically depicts example placements of barcode regions.
FIG. 3 depicts example sequences of two forked adapters ligated to opposite ends of a target polynucleotide. Full-length sequence disclosed as SEQ ID NO: 35.
FIG. 4 is a schematic example method used to generate a forked adapter described in Example 1.
FIG. 5 is a schematic example of a capsule within a capsule described in Example 2.
FIG. 6 is a schematic example of capsules within a capsule described in Example 3.
FIG. 7 is a schematic example of a product (or intermediate) that may be generated according to methods of Example 4.
FIGS. 8a-c depict example sequences described in Example 4.
FIGS. 9a-j depict example sequences described in Example 5.
FIGS. 10a-e depict example sequences described in Example 6.
FIGS. 11a-d schematically depict methods and structures described in Example 7.
FIG. 12 schematically depicts the production capsules via an example flow-focusing method.
FIG. 13 schematically depicts the production of capsules within capsules via an example flow-focusing method.
FIGS. 14a-e schematically depict methods and structures described in Example 8.
FIGS. 15a-e schematically depict methods and structures described in Example 9.
FIG. 16 schematically depicts methods and structures described in Example 10.
FIG. 17 schematically depicts a capsule within a capsule described in Example 11.
FIG. 18 schematically depicts capsules within a capsule described in Example 12.
FIGS. 19a-e depict example sequences described in Example 13. FIG. 19f describes example methods and structures described in Example 13.
FIG. 20 schematically depicts a capsule within a capsule described in Example 14.
FIGS. 21a-c schematically depict methods and structures described in Example 15.
FIG. 22 schematically depicts a capsule within a capsule described in Example 16.
FIG. 23 schematically depicts a capsule within a capsule described in Example 17.
DETAILED DESCRIPTION
While various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.
This disclosure provides methods, compositions, systems, and kits for the generation of polynucleotide barcodes and the use of such polynucleotide barcodes. Such polynucleotide barcodes may be used for any suitable application. In some cases, the polynucleotide barcodes provided in this disclosure may be used in next generation sequencing reactions. Next generation sequencing reactions include the sequencing of whole genomes, detection of specific sequences such as single nucleotide polymorphisms (SNPs) and other mutations, detection of nucleic acid (e.g., deoxyribonucleic acid) insertions, and detection of nucleic acid deletions.
Utilization of the methods, compositions, systems, and kits described herein may incorporate, unless otherwise indicated, any conventional techniques of organic chemistry, polymer technology, microfluidics, molecular biology, recombinant techniques, cell biology, biochemistry, and immunology. Such conventional techniques include well and microwell construction, capsule generation, generation of emulsions, spotting, microfluidic device construction, polymer chemistry, restriction digestion, ligation, cloning, polynucleotide sequencing, and polynucleotide sequence assembly. Specific, non-limiting, illustrations of suitable techniques are described throughout this disclosure. However, equivalent procedures may also be utilized. Descriptions of certain techniques may be found in standard laboratory manuals, such as Genome Analysis: A Laboratory Manual Series (Vols. I-IV), Using Antibodies: A Laboratory Manual, Cells: A Laboratory Manual, PCR Primer: A Laboratory Manual, and Molecular Cloning: A Laboratory Manual (all from Cold Spring Harbor Laboratory Press), and “Oligonucleotide Synthesis: A Practical Approach” 1984, IRL Press London, all of which are herein incorporated in their entirety by reference for all purposes.
I. DEFINITIONS
The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
As used herein, the singular forms “a,” “an,” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms “including,” “includes,” “having,” “has,” “with,” “such as,” or variants thereof, are used in either the specification and/or the claims, such terms are not limiting and are intended to be inclusive in a manner similar to the term “comprising”.
The term “about,” as used herein, generally refers to a range that is 15% greater than or less than a stated numerical value within the context of the particular usage. For example, “about 10” would include a range from 8.5 to 11.5.
The term “barcode,” as used herein, generally refers to a label that may be attached to an analyte to convey information about the analyte. For example, a barcode may be a polynucleotide sequence attached to fragments of a target polynucleotide contained within a particular partition. This barcode may then be sequenced with the fragments of the target polynucleotide. The presence of the same barcode on multiple sequences may provide information about the origin of the sequence. For example, a barcode may indicate that the sequence came from a particular partition and/or a proximal region of a genome. This may be particularly useful for sequence assembly when several partitions are pooled before sequencing.
The term “bp,” as used herein, generally refers to an abbreviation for “base pairs”.
The term “microwell,” as used herein, generally refers to a well with a volume of less than 1 mL. Microwells may be made in various volumes, depending on the application. For example, microwells may be made in a size appropriate to accommodate any of the partition volumes described herein.
The term “partition,” as used herein, may be a verb or a noun. When used as a verb (e.g., “to partition,” or “partitioning”), the term generally refers to the fractionation (e.g., subdivision) of a species or sample (e.g., a polynucleotide) between vessels that can be used to sequester one fraction (or subdivision) from another. Such vessels are referred to using the noun “partition.” Partitioning may be performed, for example, using microfluidics, dilution, dispensing, and the like. A partition may be, for example, a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a bead, a surface of a bead in dilute solution, or any other suitable container for sequestering one fraction of a sample from another. A partition may also comprise another partition.
The terms “polynucleotide” or “nucleic acid,” as used herein, generally refer to molecules comprising a plurality of nucleotides. Exemplary polynucleotides include deoxyribonucleic acids, ribonucleic acids, and synthetic analogues thereof, including peptide nucleic acids.
The term “species,” as used herein, generally refers to any substance that can be used with the methods, compositions, systems, devices, and kits of this disclosure. Examples of species include reagents, analytes, cells, chromosomes, tagging molecules or groups of molecules, barcodes, and any sample comprising any of these species. Any suitable species may be used, as more fully discussed elsewhere in this disclosure.
II. POLYNUCLEOTIDE BARCODING
Certain applications, for example polynucleotide sequencing, may rely on unique identifiers (“barcodes”) to identify the origin of a sequence and, for example, to assemble a larger sequence from sequenced fragments. Therefore, it may be desirable to add barcodes to polynucleotide fragments before sequencing. Barcodes may be of a variety of different formats, including polynucleotide barcodes. Depending upon the specific application, barcodes may be attached to polynucleotide fragments in a reversible or irreversible manner. Additionally, barcodes may allow for identification and/or quantification of individual polynucleotide fragments during sequencing.
Barcodes may be loaded into partitions so that one or more barcodes are introduced into a particular partition. In some cases, each partition may contain a different set of barcodes. This may be accomplished by directly dispensing the barcodes into the partitions, or by placing the barcodes within a partition that is contained within another partition.
The barcodes may be loaded into the partitions at an expected or predicted ratio of barcodes per species to be barcoded (e.g., polynucleotide fragment, strand of polynucleotide, cell, etc.). In some cases, the barcodes are loaded into partitions such that about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the barcodes are loaded into partitions such that more than about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species. In some cases, the barcodes are loaded in the partitions so that less than about 0.0001, 0.001, 0.1, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, 100, 500, 1000, 5000, 10000, or 200000 barcodes are loaded per species.
When more than one barcode is present per polynucleotide fragment, such barcodes may be copies of the same barcode, or may be different barcodes. For example, the attachment process may be designed to attach multiple identical barcodes to a single polynucleotide fragment, or multiple different barcodes to the polynucleotide fragment.
The methods provided herein may comprise loading a partition with the reagents necessary for the attachment of barcodes to polynucleotide fragments. In the case of ligation reactions, reagents including restriction enzymes, ligase enzymes, buffers, adapters, barcodes and the like may be loaded into a partition. In the case of barcoding by amplification, reagents including primers, DNA polymerases, dNTPs, buffers, barcodes and the like may be loaded into a partition. In the case of transposon-mediated barcoding (e.g., NEXTERA), reagents including a transposome (i.e., transposase and transposon end complex), buffers, and the like may be loaded into a partition. In the case of MALBAC-mediated barcoding, reagents including a MALBAC primer, buffers, and the like may be loaded into a partition. As described throughout this disclosure, these reagents may be loaded directly into the partition, or via another partition.
Barcodes may be ligated to a polynucleotide fragment using sticky or blunt ends. Barcoded polynucleotide fragments may also be generated by amplifying a polynucleotide fragment with primers comprising barcodes. In some cases, MALBAC amplification of the polynucleotide fragment may be used to generate a barcoded polynucleotide fragment. A primer used for MALBAC may or may not comprise a barcode. In cases where a MALBAC primer does not comprise a barcode, the barcode may be added to MALBAC amplification products by other amplification methods, such as, for example, PCR. Barcoded polynucleotide fragments may also be generated using transposon-mediated methods. As with any other species discussed in this disclosure, these modules may be contained within the same or different partitions, depending on the needs of the assay or process.
In some cases, barcodes may be assembled combinatorially, from smaller components designed to assemble in a modular format. For example, three modules, 1A, 1B, and 1C may be combinatorially assembled to produce barcode 1ABC. Such combinatorial assembly may significantly reduce the cost of synthesizing a plurality of barcodes. For example, a combinatorial system consisting of 3 A modules, 3 B modules, and 3 C modules may generate 3*3*3=27 possible barcode sequences from only 9 modules.
In some cases, as further described elsewhere in this disclosure, barcodes may be combinatorially assembled by mixing two oligonucleotides and hybridizing them to produce annealed or partially annealed oligonucleotides (e.g., forked adapters). These barcodes may comprise an overhang of one or more nucleotides, in order to facilitate ligation with polynucleotide fragments that are to be barcoded. In some cases, the 5′ end of the antisense strand may be phosphorylated in order to ensure double-stranded ligation. Using this approach, different modules may be assembled by, for example, mixing oligonucleotides A and B, A and C, A and D, B and C, B, and D, and so on. As described in more detail elsewhere in this disclosure, the annealed oligonucleotides may also be synthesized as a single molecule with a hairpin loop that may be cut after ligation to the polynucleotide to be barcoded.
As described in more detail elsewhere in this disclosure, attachment of polynucleotides to each other may rely on hybridization-compatible overhangs. For example, the hybridization between A and T is often used to ensure ligation compatibility between fragments. In some cases, an A overhang may be created by treatment with an enzyme, such as a Taq polymerase. In some cases, a restriction enzyme may be used to create a cleavage product with a single base 3′ overhang which may be, for example, A or T. Examples of restriction enzymes that leave a single base 3′ overhang include MnII, HphI, Hpy188I, HpyAV, HpyCH4III, MboII, BciVI, BmrI, AhdI, and XcmI. In other cases, different overhangs (e.g., 5′ overhangs, overhangs of greater than a single base) may be generated by restriction enzymes. Additional restriction enzymes that may be used to generate overhangs include BfuC1, TaqαI, BbVI, Bcc1, BceA1, BcoDI, BsmAI, and BsmFI.
III. GENERATION OF PARTITIONED BARCODE LIBRARIES
In some cases, this disclosure provides methods for the generation of partitioned barcode libraries and libraries produced according to such methods. In some cases, the methods provided herein combine random synthesis of DNA sequences, separation into partitions, amplification of separated sequences, and isolation of amplified separated sequences to provide a library of barcodes contained within partitions.
a. Random Synthesis of Polynucleotide Barcodes
In some cases, the methods described herein utilize random methods of polynucleotide synthesis, including random methods of DNA synthesis. During random DNA synthesis, any combination of A, C, G, and/or T may be added to a coupling step so that each type of base in the coupling step is coupled to a subset of the product. If A, C, G, and T are present at equivalent concentrations, approximately one-quarter of the product will incorporate each base. Successive coupling steps, and the random nature of the coupling reaction, enable the generation of 4n possible sequences, where n is the number of bases in the polynucleotide. For example, a library of random polynucleotides of length 6 could have a diversity of 46=4,096 members, while a library of length 10 would have diversity of 1,048,576 members. Therefore, very large and complex libraries can be generated. These random sequences may serve as barcodes.
Any suitable synthetic bases may also be used with the invention. In some cases, the bases included in each coupling step may be altered in order to synthesize a preferred product. For example, the number of bases present in each coupling step may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In some cases, the number of bases present in each coupling step may be at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more. In some cases, the number of bases present in each coupling step may be less than 2, 3, 4, 5, 6, 7, 8, 9, or 10.
The concentration of the individual bases may also be altered in order to synthesize the preferred product. For example, any base may be present at a concentration of about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base. In some cases, any base may be present at a concentration of at least about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base. In some cases, any base may be present at a concentration of less than about 0.1, 0.5, 1, 5, or 10-fold the concentration of another base.
The length of the random polynucleotide sequence may be any suitable length, depending on the application. In some cases, the length of the random polynucleotide sequence may be 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides. In some cases, the length of the random polynucleotide sequence may be at least 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleotides. In some cases, the length of the random polynucleotide sequence may be less than 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides.
In some cases, the library is defined by the number of members. In some cases, a library may comprise about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*1011, or 1.09951*1012 members. In some cases, a library may comprise at least about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*10, or 1.09951*1012 members. In some cases, a library may comprise less than about 256, 1024, 4096, 16384, 65536, 262144, 1048576, 4194304, 16777216, 67108864, 268435456, 1073741824, 4294967296, 17179869184, 68719476736, 2.74878*10, or 1.09951*1012 members. In some cases, the library is a barcode library. In some cases, a barcode library may comprise at least about 1000, 10000, 100000, 1000000, 2500000, 5000000, 10000000, 25000000, 50000000, or 100000000 different barcode sequences.
The random barcode libraries may also comprise other polynucleotide sequences. In some cases, these other polynucleotide sequences are non-random in nature and include, for example, primer binding sites, annealing sites for the generation of forked adapters, immobilization sequences, and regions that enable annealing with a target polynucleotide sequence, and thus barcoding of the polynucleotide sequence.
b. Separation of Polynucleotides into Partitions
After synthesis of polynucleotides comprising random barcode sequences, the polynucleotides are partitioned into separate compartments to generate a library of partitioned polynucleotides comprising barcode sequences. Any suitable method of separation and any suitable partition or partitions within partitions may be used.
In some cases, partitioning is performed by diluting the mixture of polynucleotides comprising random barcode sequences such that a particular volume of the dilution contains, on average, less than a single polynucleotide. The particular volume of the dilution may then be transferred to a partition. In any plurality of partitions, each partition is therefore likely to have one or zero polynucleotide molecules.
In some cases a dilution may be performed such that each partition comprises about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, or more molecules. In some cases a dilution may be performed such that each partition comprises at least about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, or more molecules. In some cases a dilution may be performed such that each partition comprises less than about 0.001, 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, or 2 molecules.
In some cases, about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules. In some cases, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules. In some cases, less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise the specified number of molecules.
In some cases, about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides. In some cases, at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides. In some cases, less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions comprise one or fewer polynucleotides.
In some cases, a partition is a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a surface of a bead, or any other suitable container for sequestering one fraction of a sample from another. In cases where a partition includes a bead, a primer for amplification may be attached to the bead. Partitions are described in greater detail elsewhere in this disclosure.
c. Amplification of Partitioned Polynucleotides
The polynucleotides partitioned as described above are then amplified in order to generate sufficient material for barcoding of a target polynucleotide sequence. Any suitable method of amplification may be utilized, including polymerase chain reaction (PCR), ligase chain reaction (LCR), helicase-dependent amplification, linear after the exponential PCR (LATE-PCR), asymmetric amplification, digital PCR, degenerate oligonucleotide primer PCR (DOP-PCR), primer extension pre-amplification PCR (PEP-PCR), ligation mediated PCR, rolling circle amplification, multiple displacement amplification (MDA), and single primer isothermal amplification (SPIA), emulsion PCR (ePCR), ePCR including the use of a bead, ePCR including the use of a hydrogel, multiple annealing and looping-based amplification cycles (MALBAC), and combinations thereof. MALBAC methods are described, for example, in Zong et al., Science, 338(6114), 1622-1626 (2012), which is incorporated herein by reference, in its entirety.
In some cases, amplification methods that generate single-stranded product (e.g., asymmetric amplification, SPIA, and LATE-PCR) may be preferred, for example. In some cases, amplification methods that generate double-stranded products (e.g., standard PCR) may be preferred. In some cases, an amplification method will exponentially amplify the partitioned polynucleotide. In some cases, an amplification method will linearly amplify the partitioned polynucleotide. In some cases, an amplification method will first exponentially and then linearly amplify a polynucleotide. Moreover, a single type of amplification may be used to amplify polynucleotides or amplification may be completed with sequential steps of different types of amplification. For example, ePCR may be combined with further rounds of ePCR or may be combined with a different type of amplification.
Amplification is performed until a suitable amount of polynucleotide comprising a barcode is produced. In some cases, amplification may be performed for 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, or more cycles. In some cases, amplification may be performed for at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, or more cycles. In some cases, amplification may be performed for less than 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 cycles.
In some cases, amplification may be performed until a certain amount of polynucleotide product is produced in each partition. In some cases, amplification is performed until the amount of polynucleotide product is about 10,000,000,000; 5,000,000,000; 1,000,000,000; 500,000,000; 100,000,000; 50,000,000; 10,000,000; 5,000,000; 1,000,000; 500,000; 400,000; 300,000; 200,000; or 100,000 molecules. In some cases, amplification is performed until the amount of polynucleotide product is at least about 100,000; 200,000; 300,000; 400,000; 500,000; 1,000,000; 5,000,000; 10,000,000; 50,000,000; 100,000,000; 500,000,000; 1,000,000,000; 5,000,000,000; or 10,000,000,000 molecules. In some cases, amplification is performed until the amount of polynucleotide product is less than about 10,000,000,000; 5,000,000,000; 1,000,000,000; 500,000,000; 100,000,000; 50,000,000; 10,000,000; 5,000,000; 1,000,000; 500,000; 400,000; 300,000; 200,000; or 100,000 molecules.
d. Isolation of Partitions Comprising Amplified Sequences
As described above, in some cases polynucleotides comprising barcodes are partitioned such that each partition contains, on average, less than one polynucleotide sequence. Therefore, in some cases, a fraction of the partitions will not contain a polynucleotide and therefore cannot contain an amplified polynucleotide. Thus, it may be desirable to separate partitions comprising polynucleotides from partitions not comprising polynucleotides.
In one case, partitions comprising polynucleotides are separated from partitions not comprising polynucleotides using flow-based sorting methods capable of identifying partitions comprising polynucleotides. In some cases an indicator of the presence of a polynucleotide may be used in order to differentiate partitions comprising polynucleotides from those not comprising polynucleotides.
In some cases, a nucleic acid stain may be used to identify partition comprising polynucleotides. Exemplary stains include intercalating dyes, minor-groove binders, major groove binders, external binders, and bis-intercalators. Specific examples of such dyes include SYBR green, SYBR blue, DAPI, propidium iodide, SYBR gold, ethidium bromide, acridines, proflavine, acridine orange, acriflavine, fluorcoumanin, ellipticine, daunomycin, chloroquine, distamycin D, chromomycin, homidium, mithramycin, ruthenium polypyridyls, anthramycin, phenanthridines and acridines, ethidium bromide, propidium iodide, hexidium iodide, dihydroethidium, ethidium homodimer-1 and -2, ethidium monoazide, ACMA, indoles, imidazoles (e.g., Hoechst 33258, Hoechst 33342, Hoechst 34580 and DAPI), acridine orange (also capable of intercalating), 7-AAD, actinomycin D, LDS751, hydroxystilbamidine, SYTOX Blue, SYTOX Green, SYTOX Orange, POPO-1, POPO-3, YOYO-1, YOYO-3, TOTO-1, TOTO-3, JOJO-1, LOLO-1, BOBO-1, BOBO-3, PO-PRO-1, PO-PRO-3, BO-PRO-1, BO-PRO-3, TO-PRO-1, TO-PRO-3, TO-PRO-5, JO-PRO-1, LO-PRO-1, YO-PRO-1, YO-PRO-3, PicoGreen, OliGreen, RiboGreen, SYBR Gold, SYBR Green I, SYBR Green II, SYBR DX, SYTO-40, -41, -42, -43, -44, -45 (blue), SYTO-13, -16, -24, -21, -23, -12, -11, -20, -22, -15, -14, -25 (green), SYTO-81, -80, -82, -83, -84, -85 (orange), SYTO-64, -17, -59, -61, -62, -60, and -63 (red).
In some cases, isolation methods such as magnetic separation or sedimentation of particles may be used. Such methods may include, for example, a step of attaching a polynucleotide to be amplified, a primer corresponding to said polynucleotide to be amplified, and/or a polynucleotide product of amplification to a bead. In some cases, attachment of a polynucleotide to be amplified, primer corresponding to said polynucleotide to be amplified, and/or a polynucleotide product to a bead may be via a photolabile linker, such as, for example, PC Amino C6. In cases where a photolabile linker is used, light may be used to release a linked polynucleotide from the bead. The bead may be, for example, a magnetic bead or a latex bead. The bead may then enable separation by, for example, magnetic sorting or sedimentation. Sedimentation of latex particles may be performed, for example, by centrifugation in a liquid that is more dense than latex, such as glycerol. In some cases, density gradient centrifugation may be used.
Beads may be of uniform size or heterogeneous size. In some cases, the diameter of a bead may be about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. A bead may have a diameter of at least about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a bead may have a diameter of less than about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a bead may have a diameter of about 0.001 μm to 1 mm, 0.01 μm to 900 μm, 0.1 μm to 600 μm, 100 μm to 200 μm, 100 μm to 300 μm, 100 μm to 400 μm, 100 μm to 500 μm, 100 μm to 600 μm, 20 μm to 50 μm, 150 μm to 200 μm, 150 μm to 300 μm, or 150 μm to 400 μm.
In some cases, a differential charge between the partitions comprising polynucleotides and partitions not comprising polynucleotides may be used to isolate partitions comprising polynucleotides, for example by performing electrophoresis or dielectrophoresis on the partitions.
In some cases, selective swelling or shrinking of partitions, based on differences in the osmotic pressures, may be used to identify particles comprising polynucleotides. In some instances, partitions comprising polynucleotides may be isolated by flow fractionation, solvent extraction, differential melting (e.g., with nucleic acid probes), or freezing.
Isolation of partitions comprising polynucleotides provides a library of partitioned polynucleotide barcodes with significant diversity while incurring only a one-time bulk synthesis expense.
IV. GENERATION OF ADAPTERS COMPRISING BARCODES
The barcodes described in this disclosure can have a variety of structures. In some cases, barcodes of this disclosure are a part of an adapter. Generally, an “adapter” is a structure used to enable attachment of a barcode to a target polynucleotide. An adapter may comprise, for example, a barcode, polynucleotide sequence compatible for ligation with a target polynucleotide, and functional sequences such as primer binding sites and immobilization regions.
In some cases, an adapter is a forked adapter. An example of a forked adapter is schematically depicted in FIG. 1. With reference to FIG. 1, two copies of a forked adapter structure 106 are depicted on opposite sides of a target polynucleotide 105. Each forked adapter comprises a first immobilization region 101, a second immobilization region 102, a first sequencing primer region 103, a second sequencing primer region 104 and a pair of partially complementary regions (within 103 and 104) that anneal to each other. Either the sequencing primer regions or immobilization regions may be used to immobilize the barcoded polynucleotides, for example, onto the surface of a bead. The sequencing primer regions may be used, for example, as annealing sites for sequencing primers. In some cases, an overhang may be designed to enable compatibility with a target sequence. In FIG. 1, the pair of annealed polynucleotides 103 and 104 have a 3′-T overhang, which is compatible with the 3′-A overhang on the target polynucleotide 105. A barcode may be included in any suitable portion of a forked adapter. After attachment of the forked adapter comprising the barcode to the target sequence 105, the sequencing primer regions 103 and 104 can be used to sequence the target polynucleotide. Another example of a forked adapter structure includes those used in Illumina™ library preparations and NEBNext® Multiplex Oligos for Illumina available from New England Biolabs™. Examples of non-forked adapters include those disclosed in Merriman et al., Electrophoresis, 33(23) 3397-3417 (2012), which is incorporated herein by reference, in its entirety.
FIG. 2 illustrates three schematic examples of placement of barcode regions within the forked adapter depicted in FIG. 1. In one example, a barcode 205 (BC1) is placed within the first immobilization region 201 or between the first immobilization region 201 and the first sequencing primer region 203. In another example, a barcode 206 (BC2) is placed within or adjacent to the first sequencing primer region 203. In yet another example, a barcode 207 (BC3) is placed within the second immobilization region 202 or between the second immobilization region 202 and the second sequencing primer region 204. Although FIG. 2 depicts barcodes on both ends of the target sequence, this is not necessary, as only one barcode per target sequence is sufficient for some applications. However, as described elsewhere in this disclosure, more than one barcode per target sequence may also be used.
FIG. 3 provides exemplary sequences (SEQ ID NO: 1 and SEQ ID NO: 22) of two forked adapters ligated to opposite ends of a target polynucleotide (NNN) and shows barcode regions of each forked adapter at the sequence level (bolded, nucleotides 30-37, 71-77, 81-87, and 122-129). In FIG. 3, nucleotides 1-29 represent an immobilization region of the first forked adapter, nucleotides 38-70 represent a sequencing primer region of the first forked adapter, nucleotides 78-80 (NNN) represent a target polynucleotide of arbitrary length, nucleotides 88-120 represent a sequencing primer region of the second forked adapter, and nucleotides 129-153 represent an immobilization region of the second forked adapter.
V. PARTITIONS
a. General Characteristics of Partitions
As described throughout this disclosure, certain methods, compositions, systems, devices, and kits of the disclosure may utilize the subdivision (partitioning) of certain species into separate partitions. A partition may be, for example, a well, a microwell, a hole, a droplet (e.g., a droplet in an emulsion), a continuous phase of an emulsion, a test tube, a spot, a capsule, a surface of a bead, or any other suitable container for sequestering one fraction of a sample or a species. Partitions may be used to contain a species for further processing. For example, if a species is a polynucleotide analyte, further processing may comprise cutting, ligating, and/or barcoding with species that are reagents. Any number of devices, systems or containers may be used to hold, support or contain partitions. In some cases, a microwell plate may be used to hold, support, or contain partitions. Any suitable microwell plate may be used, for example microwell plates having 96, 384, or 1536 wells.
Each partition may also contain, or be contained within any other suitable partition. For example, a well, microwell, hole, a surface of a bead, or a tube may comprise a droplet (e.g., a droplet in an emulsion), a continuous phase in an emulsion, a spot, a capsule, or any other suitable partition. A droplet may comprise a capsule, bead, or another droplet. A capsule may comprise a droplet, bead, or another capsule. These descriptions are merely illustrative, and all suitable combinations and pluralities are also envisioned. For example, any suitable partition may comprise a plurality of the same or different partitions. In one example, a well or microwell comprises a plurality of droplets and a plurality of capsules. In another example, a capsule comprises a plurality of capsules and a plurality of droplets. All combinations of partitions are envisioned. Table 1 shows non-limiting examples of partitions that may be combined with each other.
TABLE 1
Examples of partitions that may be combined with each other.
Well Spot Droplet Capsule
Well Well inside Spot inside Droplet Capsule
well well inside well inside well
Spot Spot inside Spot inside Droplet Capsule
well spot inside spot inside spot
Droplet Droplet Droplet Droplet Droplet
inside well inside spot inside droplet inside
capsule
Capsule
inside droplet
Capsule Capsule Capsule Capsule Capsule
inside well inside spot inside droplet inside
Spot inside Droplet capsule
capsule inside
capsule
Surface of a Bead inside Spot on bead Bead inside Bead inside
Bead well Bead inside droplet capsule
spot
Any partition described herein may comprise multiple partitions. For example, a partition may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. A partition may comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, a partition may comprise less than 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, or 50000 partitions. In some cases, each partition may comprise 2-50, 2-20, 2-10, or 2-5 partitions.
A partition may comprise any suitable species or mixture of species. For example, in some cases a partition may comprise a reagent, an analyte, a sample, a cell, and combinations thereof. A partition comprising other partitions may comprise certain species in the same partitions and certain species in different partitions. Species may be distributed between any suitable partitions, depending on the needs of the particular process. For example, any of the partitions in Table 1 may contain at least one first species and any of the partitions in Table 1 may contain at least one second species. In some cases the first species may be a reagent and the second species may be an analyte.
In some cases, a species is a polynucleotide isolated from a cell. For example, in some cases polynucleotides (e.g., genomic DNA, RNA, etc.) is isolated from a cell utilizing any suitable method (e.g., a commercially available kit). The polynucleotide may be quantified. The quantified polynucleotide may then be partitioned into a plurality of partitions as described herein. The partitioning of the polynucleotide may be performed at a predetermined coverage amount, according to the quantification and the needs of the assay. In some cases, all or most (e.g., at least 50%, 60%, 70%, 80%, 90%, or 95%) of the partitions do not comprise polynucleotides that overlap, such that separate mixtures of non-overlapping fragments are formed across the plurality of partitions. The partitioned polynucleotides may then be treated according to any suitable method known in the art or described in this disclosure. For example, the partitioned polynucleotides may be fragmented, amplified, barcoded, and the like.
Species may be partitioned using a variety of methods. For example, species may be diluted and dispensed across a plurality of partitions. A terminal dilution of a medium comprising species may be performed such that the number of partitions exceeds the number of species. Dilution may also be used prior to forming an emulsion or capsules, or prior to spotting a species on a substrate. The ratio of the number of species to the number of partitions may be about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000. The ratio of the number of species to the number of partitions may be at least about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000. The ratio of the number of species to the number of partitions may be less than about 0.1, 0.5, 1, 2, 4, 8, 10, 20, 50, 100, or 1000. The ratio of the number of species to the number of partitions may range from about 0.1-10, 0.5-10, 1-10, 2-10, 10-100, 100-1000, or more.
Partitioning may also be performed using piezoelectric droplet generation (e.g., Bransky et al., Lab on a Chip, 2009, 9, 516-520) or surface acoustic waves (e.g., Demirci and Montesano, Lab on a Chip, 2007, 7, 1139-1145).
The number of partitions employed may vary depending on the application. For example, the number of partitions may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000, or more. The number of partitions may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000. The number of partitions may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
The number of different barcodes or different sets of barcodes that are partitioned may vary depending upon, for example, the particular barcodes to be partitioned and/or the application. Different sets of barcodes may be, for example, sets of identical barcodes where the identical barcodes differ between each set. Or different sets of barcodes may be, for example, sets of different barcodes, where each set differs in its included barcodes. For example, about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes or different sets of barcodes may be partitioned. In some examples, at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes or different sets of barcodes may be partitioned. In some examples, less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes or different sets of barcodes may be partitioned. In some examples, about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes may be partitioned.
Barcodes may be partitioned at a particular density. For example, barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 barcodes per partition.
Barcodes may be partitioned such that identical barcodes are partitioned at a particular density. For example, identical barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more identical barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 identical barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 identical barcodes per partition.
Barcodes may be partitioned such that different barcodes are partitioned at a particular density. For example, different barcodes may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition. Barcodes may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, 100000000, or more different barcodes per partition. Barcodes may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200,000, 300,000, 400,000, 500,000, 600,000, 700,000, 800,000, 900,000, 1000000, 2000000, 3000000, 4000000, 5000000, 6000000, 7000000, 8000000, 9000000, 10000000, 20000000, 50000000, or 100000000 different barcodes per partition. Barcodes may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, 100000-1000000, 10000-1000000, 10000-10000000, or 10000-100000000 different barcodes per partition.
The number of partitions employed to partition barcodes may vary, for example, depending on the application and/or the number of different barcodes to be partitioned. For example, the number of partitions employed to partition barcodes may be about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, or 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100,000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1,000,000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more. The number of partitions employed to partition barcodes may be at least about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, 20000000 or more. The number of partitions employed to partition barcodes may be less than about 5, 10, 50, 100, 250, 500, 750, 1000, 1500, 2000, 2500, 5000, 7500, 10,000, 20000, 30000, 40000, 50000, 60000, 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, 2000000, 3000000, 4000000, 5000000, 10000000, or 20000000. The number of partitions employed to partition barcodes may be about 5-10000000, 5-5000000, 5-1,000,000, 10-10,000, 10-5,000, 10-1,000, 1,000-6,000, 1,000-5,000, 1,000-4,000, 1,000-3,000, or 1,000-2,000.
As described above, different barcodes or different sets of barcodes (e.g., each set comprising a plurality of identical barcodes or different barcodes) may be partitioned such that each partition comprises a different barcode or different barcode set. In some cases, each partition may comprise a different set of identical barcodes. Where different sets of identical barcodes are partitioned, the number of identical barcodes per partition may vary. For example, about 100,000 or more different sets of identical barcodes may be partitioned across about 100,000 or more different partitions, such that each partition comprises a different set of identical barcodes. In each partition, the number of identical barcodes per set of barcodes may be about 1,000,000 identical barcodes. In some cases, the number of different sets of barcodes may be equal to or substantially equal to the number of partitions. Any suitable number of different barcodes or different barcode sets (including numbers of different barcodes or different barcode sets to be partitioned described elsewhere herein), number of barcodes per partition (including numbers of barcodes per partition described elsewhere herein), and number of partitions (including numbers of partitions described elsewhere herein) may be combined to generate a diverse library of partitioned barcodes with high numbers of barcodes per partition. Thus, as will be appreciated, any of the above-described different numbers of barcodes may be provided with any of the above-described barcode densities per partition, and in any of the above-described numbers of partitions.
The volume of the partitions may vary depending on the application. For example, the volume of any of the partitions described in this disclosure (e.g., wells, spots, droplets (e.g., in an emulsion), and capsules) may be about 1000 μl, 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5 μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 pL, 300 pL, 200 pL, 100 pL, 50 pL, 25 pL, 10 pL, 5 pL, 1 pL, 900 fL, 800 fL, 700 fL, 600 fL, 500 fL, 400 fL, 300 fL, 200 fL, 100 fL, 50 fL, 25 fL, 10 fL, 5 fL, 1 fL, or 0.5 fL. The volume of the partitions may be at least about 1000 μl, 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5 μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 pL, 300 pL, 200 pL, 100 pL, 50 pL, 25 pL, 10 pL, 5 pL, 1 pL, 900 fL, 800 fL, 700 fL, 600 fL, 500 fL, 400 fL, 300 fL, 200 fL, 100 fL, 50 fL, 25 fL, 10 fL, 5 fL, 1 fL, or 0.5 fL. The volume of the partitions may be less than about 1000 μl, 900 μl, 800 μl, 700 μl, 600 μl, 500 μl, 400 μl, 300 μl, 200 μl, 100 μl, 50 μl, 25 μl, 10 μl, 5 μl, 1 μl, 900 nL, 800 nL, 700 nL, 600 nL, 500 nL, 400 nL, 300 nL, 200 nL, 100 nL, 50 nL, 25 nL, 10 nL, 5 nL, 5 nL, 2.5 nL, 1 nL, 900 pL, 800 pL, 700 pL, 600 pL, 500 pL, 400 pL, 300 pL, 200 pL, 100 pL, 50 pL, 25 pL, 10 pL, 5 pL, 1 pL, 900 fL, 800 fL, 700 fL, 600 fL, 500 fL, 400 fL, 300 fL, 200 fL, 100 fL, 50 fL, 25 fL, 10 fL, 5 fL, 1 fL, or 0.5 fL. the volume of the partitions may be about 0.5 fL-5 pL, 10 pL-10 nL, 10 nL-10 μl, 10 μl-100 μl, or 100 μl to 1 mL.
There may be variability in the volume of fluid in different partitions. More specifically, the volume of different partitions may vary by at least (or at most) plus or minus 1%, 2%, 3%, 4%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400%, 500%, or 1000% across a set of partitions. For example, a well (or other partition) may comprise a volume of fluid that is at most 80% of the fluid volume within a second well (or other partition).
Particular species may also be targeted to specific partitions. For example, in some cases, a capture reagent (e.g., an oligonucleotide probe) may be immobilized or placed within a partition to capture specific species (e.g., polynucleotides). For example, a capture oligonucleotide may be immobilized on the surface of a bead in order to capture a species comprising an oligonucleotide with a complementary sequence.
Species may also be partitioned at a particular density. For example, species may be partitioned so that each partition contains about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned so that each partition contains at least about 1, 5, 10, 50, 100, 1000, 10000, 100000, 1000000 or more species per partition. Species may be partitioned so that each partition contains less than about 1, 5, 10, 50, 100, 1000, 10000, 100000, or 1000000 species per partition. Species may be partitioned such that each partition contains about 1-5, 5-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 species per partition.
Species may be partitioned such that at least one partition comprises a species that is unique within that partition. This may be true for about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for at least about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more of the partitions. This may be true for less than about 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the partitions.
a. Wells as Partitions
In some cases, wells are used as partitions. The wells may be microwells. A well may comprise a medium comprising a species or plurality of species. Species may be contained within a well in various configurations. In one example, a species is dispensed directly into a well. A species dispensed directly into a well may be overlaid with a layer that is, for example, dissolvable, meltable, or permeable. This layer may be, for example, an oil, wax, membrane, or the like. The layer may be dissolved or melted prior to or after introduction of another species into the well. The well may be sealed at any point, with a sealing layer, for example after addition of any species.
In one example, reagents for sample processing are dispensed directly into a well and overlaid with a layer that is dissolvable, meltable, or permeable. A sample comprising an analyte to be processed is introduced on top of the layer. The layer is dissolved or melted, or the analyte (or reagent) diffuses through the layer. The well is sealed and incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered.
In some cases, wells comprise other partitions. A well may comprise any suitable partition including, for example, another well, a spot, a droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like. Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
In one example, a well comprises a capsule comprising reagents for sample processing. A capsule may be loaded into a well using a liquid medium, or loaded into a well without a liquid medium (e.g., essentially dry). As described elsewhere in this disclosure, a capsule may contain one or more capsules, or other partitions. A sample comprising an analyte to be processed may be introduced into the well. The well may be sealed and a stimulus may be applied to cause release of the contents of the capsule into the well, resulting in contact between the reagents and the analyte to be processed. The well may be incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the well, the opposite configuration—i.e., reagent in the well and analyte in the capsule—is also possible.
In another example, a well comprises an emulsion and the droplets of the emulsion comprise capsules comprising reagents for sample processing. A sample comprising an analyte to be processed is contained within the droplets of the emulsion. The well is sealed and a stimulus is applied to cause release of the contents of the capsules into the droplets, resulting in contact between the reagents and the analyte to be processed. The well is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in a droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
Wells may be arranged as an array, for example a microwell array. Based on the dimensions of individual wells and the size of the substrate, the well array may comprise a range of well densities. In some cases, the well density may be 10 wells/cm2, 50 wells/cm2, 100 wells/cm2, 500 wells/cm2, 1000 wells/cm2, 5000 wells/cm2, 10000 wells/cm2, 50000 wells/cm2, or 100000 wells/cm2. In some cases, the well density may be at least 10 wells/cm2, 50 wells/cm2, 100 wells/cm2, 500 wells/cm2, 1000 wells/cm2, 5000 wells/cm2, 10000 wells/cm2, 50000 wells/cm2, or 100000 wells/cm2. In some cases, the well density may be less than 10 wells/cm2, 50 wells/cm2, 100 wells/cm2, 500 wells/cm2, 1000 wells/cm2, 5000 wells/cm2, 10000 wells/cm2, 50000 wells/cm2, or 100000 wells/cm2.
b. Spots as Partitions
In some cases, spots are used as partitions. A spot may be made, for example, by dispensing a substance on a surface. Species may be contained within a spot in various configurations. In one example, a species is dispensed directly into a spot by including the species in the medium used to form the spot. A species dispensed directly onto a spot may be overlaid with a layer that is, for example, dissolvable, meltable, or permeable. This layer may be, for example, an oil, wax, membrane, or the like. The layer may be dissolved or melted prior to or after introduction of another species onto the spot. The spot may be sealed at any point, for example after addition of any species, by an overlay.
In one example, reagents for sample processing are dispensed directly onto a spot, for example on a glass slide, and overlaid with a layer that is dissolvable, meltable, or permeable. A sample comprising an analyte to be processed is introduced on top of the layer. The layer is dissolved or melted, or the analyte (or reagent) diffuses through the layer. The spot is sealed and incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered.
As described elsewhere in this disclosure (e.g., Table 1), spots may also be arranged within a well. In some cases, a plurality of spots may be arranged within a well such that the contents of each spot do not mix. Such a configuration may be useful, for example, when it is desirable to prevent species from contacting each other. In some cases, a well may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more spots. In some cases, a well may comprise at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more spots. In some cases, a well may comprise less than 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 spots. In some cases, a well may comprise 2-4, 2-6, 2-8, 4-6, 4-8, 5-10, or 4-12 spots. Upon addition of a substance (e.g., a medium containing an analyte) to the well, the species in the spot may mix. Moreover, using separate spots to contain different species (or combinations of species) may also be useful to prevent cross-contamination of devices used to place the spots inside the well.
In some cases, spots comprise other partitions. A spot may comprise any suitable partition including, for example, another spot a droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like. Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
In one example, a spot comprises a capsule comprising reagents for sample processing. As described elsewhere in this disclosure, a capsule may contain one or more capsules, or other partitions. A sample comprising an analyte to be processed is introduced into the spot. The spot is sealed and a stimulus is applied to cause release of the contents of the capsule into the spot, resulting in contact between the reagents and the analyte to be processed. The spot is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the spot, the opposite configuration—i.e., reagent in the spot and analyte in the capsule—is also possible.
In another example, a spot comprises an emulsion and the droplets of the emulsion comprise capsules comprising reagents for sample processing. A sample comprising an analyte to be processed is contained within the droplets of the emulsion. The spot is sealed and a stimulus is applied to cause release of the contents of the capsules into the droplets, resulting in contact between the reagents and the analyte to be processed. The spot is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in a droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
Spots may be of uniform size or heterogeneous size. In some cases, the diameter of a spot may be about 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, 1 mm, 2 mm, 5 mm, or 1 cm. A spot may have a diameter of at least about 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, 1 mm, 1 mm, 2 mm, 5 mm, or 1 cm. In some cases, a spot may have a diameter of less than about 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, 1 mm, 1 mm, 2 mm, 5 mm, or 1 cm. In some cases, a spot may have a diameter of about 0.1 μm to 1 cm, 100 μm to 1 mm, 100 μm to 500 μm, 100 μm to 600 μm, 150 μm to 300 μm, or 150 μm to 400 μm.
Spots may be arranged as an array, for example a spot array. Based on the dimensions of individual spots and the size of the substrate, the spot array may comprise a range of spot densities. In some cases, the spot density may be 10 spots/cm2, 50 spots/cm2, 100 spots/cm2, 500 spots/cm2, 1000 spots/cm2, 5000 spots/cm2, 10000 spots/cm2, 50000 spots/cm2, or 100000 spots/cm2. In some cases, the spot density may be at least 10 spots/cm2, 50 spots/cm2, 100 spots/cm2, 500 spots/cm2, 1000 spots/cm2, 5000 spots/cm2, 10000 spots/cm2, 50000 spots/cm2, or 100000 spots/cm2. In some cases, the spot density may be less than 10 spots/cm2, 50 spots/cm2, 100 spots/cm2, 500 spots/cm2, 1000 spots/cm2, 5000 spots/cm2, 10000 spots/cm2, 50000 spots/cm2, or 100000 spots/cm2.
c. Emulsions as Partitions
In some cases, the droplets in an emulsion are used as partitions. An emulsion may be prepared, for example, by any suitable method, including methods known in the art. (See e.g., Weizmann et al., Nature Methods, 2006, 3(7):545-550; Weitz et al. U.S. Pub. No. 2012/0211084). In some cases, water-in-fluorocarbon emulsions may be used. These emulsions may incorporate fluorosurfactants such as oligomeric perfluorinated polyethers (PFPE) with polyethylene glycol (PEG). (Holtze et al., Lab on a Chip, 2008, 8(10):1632-1639). In some cases, monodisperse emulsions may be formed in a microfluidic flow focusing device. (Garstecki et al., Applied Physics Letters, 2004, 85(13):2649-2651).
A species may be contained within a droplet in an emulsion containing, for example, a first phase (e.g., oil or water) forming the droplet and a second (continuous) phase (e.g., water or oil). An emulsion may be a single emulsion, for example, a water-in-oil or an oil-in-water emulsion. An emulsion may be a double emulsion, for example a water-in-oil-in-water or an oil-in-water-in-oil emulsion. Higher-order emulsions are also possible. The emulsion may be held in any suitable container, including any suitable partition described in this disclosure.
In some cases, droplets in an emulsion comprise other partitions. A droplet in an emulsion may comprise any suitable partition including, for example, another droplet (e.g., a droplet in an emulsion), a capsule, a bead, and the like. Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
In one example, a droplet in an emulsion comprises a capsule comprising reagents for sample processing. As described elsewhere in this disclosure, a capsule may contain one or more capsules, or other partitions. A sample comprising an analyte to be processed is contained within the droplet. A stimulus is applied to cause release of the contents of the capsule into the droplet, resulting in contact between the reagents and the analyte to be processed. The droplet is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in a capsule and an analyte is in the droplet, the opposite configuration—i.e., reagent in the droplet and analyte in the capsule—is also possible.
The droplets in an emulsion may be of uniform size or heterogeneous size. In some cases, the diameter of a droplet in an emulsion may be about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. A droplet may have a diameter of at least about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a droplet may have a diameter of less than about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a droplet may have a diameter of about 0.001 μm to 1 mm, 0.01 μm to 900 μm, 0.1 μm to 600 μm, 100 μm to 200 μm, 100 μm to 300 μm, 100 μm to 400 μm, 100 μm to 500 μm, 100 μm to 600 μm, 150 μm to 200 μm, 150 μm to 300 μm, or 150 μm to 400 μm.
Droplets in an emulsion also may have a particular density. In some cases, the droplets are less dense than an aqueous fluid (e.g., water); in some cases, the droplets are denser than an aqueous fluid. In some cases, the droplets are less dense than a non-aqueous fluid (e.g., oil); in some cases, the droplets are denser than a non-aqueous fluid. Droplets may have a density of about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Droplets may have a density of at least about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. In other cases, droplet densities may be at most about 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Such densities can reflect the density of the capsule in any particular fluid (e.g., aqueous, water, oil, etc.)
d. Capsules as Partitions
In some cases, capsules are used as partitions. A capsule may be prepared by any suitable method, including methods known in the art, including emulsification polymerization (Weitz et al. (U.S. Pub. No. 2012/0211084)), layer-by-layer assembly with polyelectrolytes, coacervation, internal phase separation, and flow focusing. Any suitable species may be contained within a capsule. The capsule may be held in any suitable container, including any suitable partition described in this disclosure.
In some cases, capsules comprise other partitions. A capsule may comprise any suitable partition including, for example, another capsule, a droplet in an emulsion, a bead, and the like. Each partition may be present as a single partition or a plurality of partitions, and each partition may comprise the same species or different species.
In one example, an outer capsule comprises an inner capsule. The inner capsule comprises reagents for sample processing. An analyte is encapsulated in the medium between the inner capsule and the outer capsule. A stimulus is applied to cause release of the contents of the inner capsule into the outer capsule, resulting in contact between the reagents and the analyte to be processed. The outer capsule is incubated under appropriate conditions for the processing of the analyte. Processed analyte may then be recovered. While this example describes an embodiment where a reagent is in an inner capsule and an analyte in the medium between the inner capsule and the outer capsule, the opposite configuration—i.e., reagent in the medium between the inner capsule and the outer capsule, and analyte in the inner capsule—is also possible.
Capsules may be pre-formed and filled with reagents by injection. For example, the picoinjection methods described in Abate et al. (Proc. Natl. Acad. Sci. U.S.A., 2010, 107(45), 19163-19166) and Weitz et al. (U.S. Pub. No. 2012/0132288) may be used to introduce reagents into the interior of capsules described herein. Generally, the picoinjection will be performed prior to the hardening of the capsule shell, for example by injecting species into the interior of a capsule precursor, such as a droplet of an emulsion, before formation of the capsule shell.
Capsules may be of uniform size or heterogeneous size. In some cases, the diameter of a capsule may be about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. A capsule may have a diameter of at least about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a capsule may have a diameter of less than about 0.001 μm, 0.01 μm, 0.05 μm, 0.1 μm, 0.5 μm, 1 μm, 5 μm, 10 μm, 50 μm, 100 μm, 150 μm, 200 μm, 300 μm, 400 μm, 500 μm, 600 μm, 700 μm, 800 μm, 900 μm, or 1 mm. In some cases, a capsule may have a diameter of about 0.001 μm to 1 mm, 0.01 μm to 900 μm, 0.1 μm to 600 μm, 100 μm to 200 μm, 100 μm to 300 μm, 100 μm to 400 μm, 100 μm to 500 μm, 100 μm to 600 μm, 150 μm to 200 μm, 150 μm to 300 μm, or 150 μm to 400 μm.
Capsules also may have a particular density. In some cases, the capsules are less dense than an aqueous fluid (e.g., water); in some cases, the capsules are denser than an aqueous fluid. In some cases, the capsules are less dense than a non-aqueous fluid (e.g., oil); in some cases, the capsules are denser than a non-aqueous fluid. Capsules may have a density of about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Capsules may have a density of at least about 0.05 g/cm3, 0.1 g/cm3, 0.2 g/cm3, 0.3 g/cm3, 0.4 g/cm3, 0.5 g/cm3, 0.6 g/cm3, 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. In other cases, capsule densities may be at most about 0.7 g/cm3, 0.8 g/cm3, 0.81 g/cm3, 0.82 g/cm3, 0.83 g/cm3, 0.84 g/cm3, 0.85 g/cm3, 0.86 g/cm3, 0.87 g/cm3, 0.88 g/cm3, 0.89 g/cm3, 0.90 g/cm3, 0.91 g/cm3, 0.92 g/cm3, 0.93 g/cm3, 0.94 g/cm3, 0.95 g/cm3, 0.96 g/cm3, 0.97 g/cm3, 0.98 g/cm3, 0.99 g/cm3, 1.00 g/cm3, 1.05 g/cm3, 1.1 g/cm3, 1.2 g/cm3, 1.3 g/cm3, 1.4 g/cm3, 1.5 g/cm3, 1.6 g/cm3, 1.7 g/cm3, 1.8 g/cm3, 1.9 g/cm3, 2.0 g/cm3, 2.1 g/cm3, 2.2 g/cm3, 2.3 g/cm3, 2.4 g/cm3, or 2.5 g/cm3. Such densities can reflect the density of the capsule in any particular fluid (e.g., aqueous, water, oil, etc.)
1. Production of Capsules by Flow Focusing
In some cases, capsules may be produced by flow focusing. Flow focusing is a method whereby a first fluid that is immiscible with a second fluid is flowed into the second fluid. With reference to FIG. 12, a first (e.g., aqueous) fluid comprising a monomer, crosslinker, initiator, and aqueous surfactant 1201 is flowed into a second (e.g., oil) fluid comprising a surfactant and an accelerator 1202. After entering the second fluid at a T-junction in a microfluidic device 1203, a droplet of first fluid breaks off from the first fluid stream and a capsule shell begins to form 1204 due to the mixing of the monomer, crosslinker, and initiator in the first fluid and the accelerator in the second fluid. Thus, a capsule is formed. As the capsule proceeds downstream, the shell becomes thicker due to increased exposure to the accelerator. Varying the concentrations of the reagents may also be used to vary the thickness and permeability of the capsule shell.
A species, or other partition such as a droplet, may be encapsulated by, for example, including the species in the first fluid. Including the species in the second fluid may embed the species in the shell of the capsule. Of course, depending on the needs of the particular sample processing method, the phases may also be reversed—i.e., the first phase may be an oil phase and the second phase may be an aqueous phase.
2. Production of Capsules within Capsules by Flow Focusing
In some cases, capsules within capsules may be produced by flow focusing. With reference to FIG. 13, a first (e.g., aqueous) fluid comprising a capsule, monomer, crosslinker, initiator, and aqueous surfactant 1301 is flowed into a second (oil) fluid comprising a surfactant and an accelerator 1302. After entering the second fluid at a T-junction in a microfluidic device 1303, a droplet of first fluid breaks off from the first fluid stream and a second capsule shell begins to form around the capsule 1304 due to the mixing of the monomer, crosslinker, and initiator in the first fluid and the accelerator in the second fluid. Thus, a capsule within a capsule is formed. As the capsule proceeds downstream, the shell becomes thicker due to increased exposure to the accelerator. Varying the concentrations of the reagents may also be used to vary the thickness and permeability of the second capsule shell.
A species may be encapsulated by, for example, including the species in the first fluid. Including the species in the second fluid may embed the species in the second shell of the capsule. Of course, depending on the needs of the particular sample processing method, the phases may also be reversed—i.e., the first phase may be an oil phase and the second phase may be an aqueous phase.
3. Production of Capsules in Batch
In some cases, capsules may be produced in batch, using capsule precursors, such as the droplets in an emulsion. Capsule precursors may be formed by any suitable method, for example by producing an emulsion with droplets comprising a monomer, a crosslinker, an initiator, and a surfactant. An accelerator may then be added to the medium, resulting in the formation of capsules. As for the methods of flow focusing, the thickness of the shell can be varied by varying the concentrations of the reactants, and the time of exposure to the accelerator. The capsules may then be washed and recovered. As for any method described herein, a species, including other partitions, may be encapsulated within the capsule or, if suitable, within the shell.
In another example, the droplets of an emulsion may be exposed to an accelerator that is present in an outlet well during the emulsion generation process. For example, capsule precursors may be formed by any suitable method, such as the flow focusing method illustrated in FIG. 12. Rather than including the accelerator in second fluid 1202, the accelerator may be included in a medium located at the exit of the T-junction (e.g., a medium located at the far-right of the horizontal channel of FIG. 12. As the emulsion droplets (i.e., capsule precursors) exit the channel, they contact the medium comprising the accelerator (i.e., the outlet medium). If the capsule precursor has a density that is less than the density of outlet medium, the capsule precursors will rise through the medium, ensuring convectional and diffusional exposure to the accelerator and reducing the likelihood of polymerization at the outlet of the channel.
VI. SPECIES
The methods, compositions, systems, devices, and kits of this disclosure may be used with any suitable species. A species can be, for example, any substance used in sample processing, such as a reagent or an analyte. Exemplary species include whole cells, chromosomes, polynucleotides, organic molecules, proteins, polypeptides, carbohydrates, saccharides, sugars, lipids, enzymes, restriction enzymes, ligases, polymerases, barcodes, adapters, small molecules, antibodies, fluorophores, deoxynucleotide triphosphates (dNTPs), dideoxynucleotide triphosphates (ddNTPs), buffers, acidic solutions, basic solutions, temperature-sensitive enzymes, pH-sensitive enzymes, light-sensitive enzymes, metals, metal ions, magnesium chloride, sodium chloride, manganese, aqueous buffer, mild buffer, ionic buffer, inhibitors, saccharides, oils, salts, ions, detergents, ionic detergents, non-ionic detergents, oligonucleotides, nucleotides, DNA, RNA, peptide polynucleotides, complementary DNA (cDNA), double stranded DNA (dsDNA), single stranded DNA (ssDNA), plasmid DNA, cosmid DNA, chromosomal DNA, genomic DNA, viral DNA, bacterial DNA, mtDNA (mitochondrial DNA), mRNA, rRNA, tRNA, nRNA, siRNA, snRNA, snoRNA, scaRNA, microRNA, dsRNA, ribozyme, riboswitch and viral RNA, a locked nucleic acid (LNA) in whole or part, locked nucleic acid nucleotides, any other type of nucleic acid analogue, proteases, nucleases, protease inhibitors, nuclease inhibitors, chelating agents, reducing agents, oxidizing agents, probes, chromophores, dyes, organics, emulsifiers, surfactants, stabilizers, polymers, water, small molecules, pharmaceuticals, radioactive molecules, preservatives, antibiotics, aptamers, and the like. In summary, the species that are used will vary depending on the particular sample processing needs.
In some cases, a partition comprises a set of species that have a similar attribute (e.g., a set of enzymes, a set of minerals, a set of oligonucleotides, a mixture of different barcodes, a mixture of identical barcodes). In other cases, a partition comprises a heterogeneous mixture of species. In some cases, the heterogeneous mixture of species comprises all components necessary to perform a particular reaction. In some cases, such mixture comprises all components necessary to perform a reaction, except for 1, 2, 3, 4, 5, or more components necessary to perform the reaction. In some cases, such additional components are contained within a different partition or within a solution within or surrounding a partition.
A species may be naturally-occurring or synthetic. A species may be present in a sample obtained using any methods known in the art. In some cases, a sample may be processed before analyzing it for an analyte.
A species may be obtained from any suitable location, including from organisms, whole cells, cell preparations and cell-free compositions from any organism, tissue, cell, or environment. A species may be obtained from environmental samples, biopsies, aspirates, formalin fixed embedded tissues, air, agricultural samples, soil samples, petroleum samples, water samples, or dust samples. In some instances, a species may be obtained from bodily fluids which may include blood, urine, feces, serum, lymph, saliva, mucosal secretions, perspiration, central nervous system fluid, vaginal fluid, or semen. Species may also be obtained from manufactured products, such as cosmetics, foods, personal care products, and the like. Species may be the products of experimental manipulation including, recombinant cloning, polynucleotide amplification, polymerase chain reaction (PCR) amplification, purification methods (such as purification of genomic DNA or RNA), and synthesis reactions.
In some cases, a species may quantified by mass. A species may be provided in a mass of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng, 1 μg, 5 μg, 10 μg, 15 μg, or 20 μg. A species may be provided in a mass of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng, 1 μg, 5 μg, 10 μg, 15 μg, or 20 μg. A species may be provided in a mass of less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000 ng 1 μg, 5 μg, 10 μg, 15 μg, or 20 μg. A species may be provided in a mass ranging from about 1-10, 10-50, 50-100, 100-200, 200-1000, 1000-10000 ng, 1-5 μg, or 1-20 μg. As described elsewhere in this disclosure, if a species is a polynucleotide, amplification may be used to increase the quantity of a polynucleotide.
Polynucleotides may also be quantified as “genome equivalents.” A genome equivalent is an amount of polynucleotide equivalent to one haploid genome of an organism from which the target polynucleotide is derived. For example, a single diploid cell contains two genome equivalents of DNA. Polynucleotides may be provided in an amount ranging from about 1-10, 10-50, 50-100, 100-1000, 1000-10000, 10000-100000, or 100000-1000000 genome equivalents. Polynucleotides may be provided in an amount of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents. Polynucleotides may be provided in an amount less than about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 50, 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10000, 20000, 30000, 40000, 50000, 60000 70000, 80000, 90000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, or 1000000 genome equivalents.
Polynucleotides may also be quantified by the amount of sequence coverage provided. The amount of sequence coverage refers to the average number of reads representing a given nucleotide in a reconstructed sequence. Generally, the greater the number of times a region is sequenced, the more accurate the sequence information obtained. Polynucleotides may be provided in an amount that provides a range of sequence coverage from about 0.1×-10×, 10×-50×, 50×-100×, 100×-200×, or 200×-500×. Polynucleotides may be provided in an amount that provides at least about 0.1×, 0.2×, 0.3×, 0.4×, 0.5×, 0.6×, 0.7×, 0.8×, 0.9×, 1.0×, 5×, 10×, 25×, 50×, 100×, 125×, 150×, 175×, or 200× sequence coverage. Polynucleotides may be provided in an amount that provides less than about 0.2×, 0.3×, 0.4×, 0.5×, 0.6×, 0.7×, 0.8×, 0.9×, 1.0×, 5×, 10×, 25×, 50×, 100×, 125×, 150×, 175×, or 200× sequence coverage.
In some cases, species are introduced into a partition either before or after a particular step. For example, a lysis buffer reagent may be introduced into a partition following partitioning of a cellular sample into the partitions. In some cases, reagents and/or partitions comprising reagents are introduced sequentially such that different reactions or operations occur at different steps. The reagents (or partitions comprising reagents) may be also be loaded at steps interspersed with a reaction or operation step. For example, capsules comprising reagents for fragmenting molecules (e.g., nucleic acids) may be loaded into a well, followed by a fragmentation step, which may be followed by loading of capsules comprising reagents for ligating barcodes (or other unique identifiers, e.g., antibodies) and subsequent ligation of the barcodes to the fragmented molecules.
VII. PROCESSING OF ANALYTES AND OTHER SPECIES
In some cases, the methods, compositions, systems, devices, and kits of this disclosure may be used to process a sample containing a species, for example an analyte. Any suitable process can be performed.
a. Fragmentation of Target Polynucleotides
In some cases, the methods, compositions, systems, devices, and kits of this disclosure may be used for polynucleotide fragmentation. Fragmentation of polynucleotides is used as a step in a variety of methods, including polynucleotide sequencing. The size of the polynucleotide fragments, typically described in terms of length (quantified by the linear number of nucleotides per fragment), may vary depending on the source of the target polynucleotide, the method used for fragmentation, and the desired application. A single fragmentation step or a plurality of fragmentation steps may be used.
Fragments generated using the methods described herein may be about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides in length. Fragments generated using the methods described herein may be at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides in length. Fragments generated using the methods described herein may be less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides in length.
Fragments generated using the methods described herein may have a mean or median length of about 1-10, 10-20, 20-50, 50-100, 50-200, 100-200, 200-300, 300-400, 400-500, 500-1000, 1000-5000, 5000-10000, 10000-100000, 100000-250000, or 250000-500000 nucleotides.
Fragments generated using the methods described herein may have a mean or median length of at least about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, or more nucleotides. Fragments generated using the methods described herein may have a mean or median length of less than about 10, 20, 100, 200, 300, 400, 500, 1000, 5000, 10000, 100000, 250000, 500000, nucleotides.
Numerous fragmentation methods are known in the art. For example, fragmentation may be performed through physical, mechanical or enzymatic methods. Physical fragmentation may include exposing a target polynucleotide to heat or to UV light. Mechanical disruption may be used to mechanically shear a target polynucleotide into fragments of the desired range. Mechanical shearing may be accomplished through a number of methods known in the art, including repetitive pipetting of the target polynucleotide, sonication (e.g., using ultrasonic waves), cavitation and nebulization. Target polynucleotides may also be fragmented using enzymatic methods. In some cases, enzymatic digestion may be performed using enzymes such as using restriction enzymes.
While the methods of fragmentation described in the preceding paragraph, and in some paragraphs of the disclosure, are described with reference to “target” polynucleotides, this is not meant to be limiting, above or anywhere else in this disclosure. Any method of fragmentation described herein, or known in the art, can be applied to any polynucleotide used with the invention. In some cases, this polynucleotide may be a target polynucleotide, such as a genome.
In other cases, this polynucleotide may be a fragment of a target polynucleotide which one wishes to further fragment. In still other cases, still further fragments may be still further fragmented. Any suitable polynucleotide may be fragmented according the methods described herein.
Restriction enzymes may be used to perform specific or non-specific fragmentation of target polynucleotides. The methods of the present disclosure may use one or more types of restriction enzymes, generally described as Type I enzymes, Type II enzymes, and/or Type III enzymes. Type II and Type III enzymes are generally commercially available and well known in the art. Type II and Type III enzymes recognize specific sequences of nucleotide base pairs within a double stranded polynucleotide sequence (a “recognition sequence” or “recognition site”). Upon binding and recognition of these sequences, Type II and Type III enzymes cleave the polynucleotide sequence. In some cases, cleavage will result in a polynucleotide fragment with a portion of overhanging single stranded DNA, called a “sticky end.” In other cases, cleavage will not result in a fragment with an overhang, creating a “blunt end.” The methods of the present disclosure may comprise use of restriction enzymes that generate either sticky ends or blunt ends.
Restriction enzymes may recognize a variety of recognition sites in the target polynucleotide. Some restriction enzymes (“exact cutters”) recognize only a single recognition site (e.g., GAATTC). Other restriction enzymes are more promiscuous, and recognize more than one recognition site, or a variety of recognition sites. Some enzymes cut at a single position within the recognition site, while others may cut at multiple positions. Some enzymes cut at the same position within the recognition site, while others cut at variable positions.
The present disclosure provides method of selecting one or more restriction enzymes to produce fragments of a desired length. Polynucleotide fragmentation may be simulated in silico, and the fragmentation may be optimized to obtain the greatest number or fraction of polynucleotide fragments within a particular size range, while minimizing the number or fraction of fragments within undesirable size ranges. Optimization algorithms may be applied to select a combination of two or more enzymes to produce the desired fragment sizes with the desired distribution of fragments quantities.
A polynucleotide may be exposed to two or more restriction enzymes simultaneously or sequentially. This may be accomplished by, for example, adding more than one restriction enzyme to a partition, or by adding one restriction enzyme to a partition, performing the digestion, deactivating the restriction enzyme (e.g., by heat treatment) and then adding a second restriction enzyme. Any suitable restriction enzyme may be used alone, or in combination, in the methods presented herein.
In some cases, a species is a restriction enzyme that is a “rare-cutter.” The term “rare-cutter enzyme,” as used herein, generally refers to an enzyme with a recognition site that occurs only rarely in a genome. The size of restriction fragments generated by cutting a hypothetical random genome with a restriction enzyme may be approximated by 4N, where N is the number of nucleotides in the recognition site of the enzyme. For example, an enzyme with a recognition site consisting of 7 nucleotides would cut a genome once every 47 bp, producing fragments of about 16,384 bp. Generally rare-cutter enzymes have recognition sites comprising 6 or more nucleotides. For example, a rare cutter enzyme may have a recognition site comprising or consisting of 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 nucleotides. Examples of rare-cutter enzymes include NotI (GCGGCCGC), XmaIII (CGGCCG), SstII (CCGCGG), SalI (GTCGAC), NruI (TCGCGA), NheI (GCTAGC), Nb.BbvCI (CCTCAGC), BbvCI (CCTCAGC), AscI (GGCGCGCC), AsiSI (GCGATCGC), FseI (GGCCGGCC), PacI (TTAATTAA), PmeI (GTTTAAAC), SbfI (CCTGCAGG), SgrAI (CRCCGGYG), SwaI (ATTTAAAT), BspQI (GCTCTTC), SapI (GCTCTTC), SfiI (GGCCNNNNNGGCC) (SEQ ID NO: 27), CspCI (CAANNNNNGTGG) (SEQ ID NO: 28), AbsI (CCTCGAGG), CciNI (GCGGCCGC), FspAI (RTGCGCAY), MauBI (CGCGCGCG), MreI (CGCCGGCG), MssI (GTTTAAAC), PalAI (GGCGCGCC), RgaI (GCGATCGC), RigI (GGCCGGCC), SdaI (CCTGCAGG), SfaAI (GCGATCGC), SgfI (GCGATCGC), SgrDI (CGTCGACG), SgsI (GGCGCGCC), SmiI (ATTTAAAT), SrfI (GCCCGGGC), Sse2321 (CGCCGGCG), Sse83871 (CCTGCAGG), LguI (GCTCTTC), PciSI (GCTCTTC), AarI (CACCTGC), AjuI (GAANNNNNNNTTGG) (SEQ ID NO: 29), AloI (GAACNNNNNNTCC) (SEQ ID NO: 30), BarI (GAAGNNNNNNTAC) (SEQ ID NO: 31), PpiI (GAACNNNNNCTC) (SEQ ID NO: 32), PsrI (GAACNNNNNNTAC) (SEQ ID NO: 33), and others.
In some cases, polynucleotides may be fragmented and barcoded at the same time. For example, a transposase (e.g., NEXTERA) may be used to fragment a polynucleotide and add a barcode to the polynucleotide.
VIII. STIMULI-RESPONSIVENESS
In some cases, stimuli may be used to trigger the release of a species from a partition. Generally, a stimulus may cause disruption of the structure of a partition, such as the wall of a well, a component of a spot, the stability of a droplet (e.g., a droplet in an emulsion), or the shell of a capsule. These stimuli are particularly useful in inducing a partition to release its contents. Because a partition may be contained within another partition, and each partition may be responsive (or not responsive) to different stimuli, stimuli-responsiveness may be employed to release the contents of one partition (e.g., a partition responsive to the stimulus) into another partition (e.g., a partition not responsive to that stimulus, or less responsive to that stimulus).
In some cases, the contents of an inner capsule may be released into the contents of an outer capsule by applying a stimulus that dissolves the inner capsule, resulting in a capsule containing a mixed sample. Of course, this embodiment is purely illustrative, and stimuli-responsiveness may be used to release the contents of any suitable partition into any other suitable partition, medium, or container (see, e.g., Table 1 for more specific examples of partitions within partitions).
Examples of stimuli that may be used include chemical stimuli, bulk changes, biological stimuli, light, thermal stimuli, magnetic stimuli, addition of a medium to a well, and any combination thereof, as described more fully below. (See, e.g., Esser-Kahn et al., (2011) Macromolecules 44: 5539-5553; Wang et al., (2009) ChemPhysChem 10:2405-2409.)
a. Chemical Stimuli and Bulk Changes
Numerous chemical triggers may be used to trigger the disruption of partitions (e.g., Plunkett et al., Biomacromolecules, 2005, 6:632-637). Examples of these chemical changes may include, but are not limited to pH-mediated changes to the integrity of a component of a partition, disintegration of a component of a partition via chemical cleavage of crosslink bonds, and triggered depolymerization of a component of a partition. Bulk changes may also be used to trigger disruption of partitions.
A change in pH of a solution, such as a decrease in pH, may trigger disruption of a partition via a number of different mechanisms. The addition of acid may cause degradation or disassembly a portion of a partition through a variety of mechanisms. Addition of protons may disassemble cross-linking of polymers in a component of a partition, disrupt ionic or hydrogen bonds in a component of a partition, or create nanopores in a component of a partition to allow the inner contents to leak through to the exterior. A change in pH may also destabilize an emulsion, leading to release of the contents of the droplets.
In some examples, a partition is produced from materials that comprise acid-degradable chemical cross-linkers, such a ketals. A decrease in pH, particular to a pH lower than 5, may induce the ketal to convert to a ketone and two alcohols and facilitate disruption of the partition. In other examples, the partitions may be produced from materials comprising one or more polyelectrolytes that are pH sensitive. A decrease in pH may disrupt the ionic- or hydrogen-bonding interactions of such partitions, or create nanopores therein. In some cases, partitions made from materials comprising polyelectrolytes comprise a charged, gel-based core that expands and contracts upon a change of pH.
Disruption of cross-linked materials comprising a partition can be accomplished through a number of mechanisms. In some examples, a partition can be contacted with various chemicals that induce oxidation, reduction or other chemical changes. In some cases, a reducing agent, such as beta-mercaptoethanol, can be used, such that disulfide bonds of a partition are disrupted. In addition, enzymes may be added to cleave peptide bonds in materials forming a partition, thereby resulting in a loss of integrity of the partition.
Depolymerization can also be used to disrupt partitions. A chemical trigger may be added to facilitate the removal of a protecting head group. For example, the trigger may cause removal of a head group of a carbonate ester or carbamate within a polymer, which in turn causes depolymerization and release of species from the inside of a partition.
In yet another example, a chemical trigger may comprise an osmotic trigger, whereby a change in ion or solute concentration in a solution induces swelling of a material used to make a partition. Swelling may cause a buildup of internal pressure such that a partition ruptures to release its contents. Swelling may also cause an increase in the pore size of the material, allowing species contained within the partition to diffuse out, and vice versa.
A partition may also be made to release its contents via bulk or physical changes, such as pressure induced rupture, melting, or changes in porosity.
b. Biological Stimuli
Biological stimuli may also be used to trigger disruption of partitions. Generally, biological triggers resemble chemical triggers, but many examples use biomolecules, or molecules commonly found in living systems such as enzymes, peptides, saccharides, fatty acids, nucleic acids and the like. For example, partitions may be made from materials comprising polymers with peptide cross-links that are sensitive to cleavage by specific proteases. More specifically, one example may comprise a partition made from materials comprising GFLGK (SEQ ID NO: 34) peptide cross links. Upon addition of a biological trigger such as the protease Cathepsin B, the peptide cross links of the shell well are cleaved and the contents of the capsule are released. In other cases, the proteases may be heat-activated. In another example, partitions comprise a component comprising cellulose. Addition of the hydrolytic enzyme chitosan serves as biologic trigger for cleavage of cellulosic bonds, depolymerization of component of the partition comprising chitosan, and release of its inner contents.
c. Thermal Stimuli
Partitions may also be induced to release their contents upon the application of a thermal stimulus. A change in temperature can cause a variety changes to a partition. A change in heat may cause melting of a partition such that a portion of the partition disintegrates, or disruption of an emulsion. In other cases, heat may increase the internal pressure of the inner components of a partition such that the partition ruptures or explodes. In still other cases, heat may transform a partition into a shrunken dehydrated state. Heat may also act upon heat-sensitive polymers used as materials to construct partitions.
In one example, a partition is made from materials comprising a thermo-sensitive hydrogel. Upon the application of heat, such as a temperature above 35 C, the hydrogel material shrinks. The sudden shrinkage of the material increases the pressure and ruptures the partition.
In some cases, a material used to produce a partition may comprise a diblock polymer, or a mixture of two polymers, with different heat sensitivities. One polymer may be particularly likely to shrink after the application of heat, while the other is more heat-stable. When heat is applied to such shell wall, the heat-sensitive polymer may shrink, while the other remains intact, causing a pore to form. In still other cases, a material used to produce a partition may comprise magnetic nanoparticles. Exposure to a magnetic field may cause the generation of heat, leading to rupture of the partition.
d. Magnetic Stimuli
Inclusion of magnetic nanoparticles in a material used to produce a partition may allow triggered rupture of the partition, as described above, as well as enable guidance of these partitions to other partitions (e.g., guidance of capsules to wells in an array). In one example, incorporation of Fe3O4 nanoparticles into materials used to produce partitions triggers rupture in the presence of an oscillating magnetic field stimulus.
e. Electrical and Light Stimuli
A partition may also be disrupted as the result of electrical stimulation. Similar to the magnetic particles described in the previous section, electrically sensitive particles can allow for both triggered rupture of partitions, as well as other functions such as alignment in an electric field or redox reactions. In one example, partitions made from materials comprising electrically sensitive material are aligned in an electric field such that release of inner reagents can be controlled. In other examples, electric fields may induce redox reactions within a partition that may increase porosity.
A light stimulus may also be used to disrupt the partitions. Numerous light triggers are possible and may include systems that use various molecules such as nanoparticles and chromophores capable of absorbing photons of specific ranges of wavelengths. For example, metal oxide coatings can be used to produce certain partitions. UV irradiation of partitions coated with SiO2/TiO2 may result in disintegration of the partition wall. In yet another example, photo switchable materials such as azobenzene groups may be incorporated in the materials used to produce the partitions. Upon the application of UV or visible light, chemicals such as these undergo a reversible cis-to-trans isomerization upon absorption of photons. In this aspect, incorporation of photo switches results in disintegration of a portion of a partition, or an increase in porosity of a portion of a partition.
f. Application of Stimuli
The devices, methods, compositions, systems, and kits of this disclosure may be used in combination with any apparatus or device that provides such trigger or stimulus. For example, if the stimulus is thermal, a device may be used in combination with a heated or thermally controlled plate, which allows heating of the wells and may induce the rupture of capsules. Any of a number of methods of heat transfer may be used for thermal stimuli, including but not limited to applying heat by radiative heat transfer, convective heat transfer, or conductive heat transfer. In other cases, if the stimulus is a biological enzyme, the enzyme may be injected into a device such that it is deposited into each well. In another aspect, if the stimulus is a magnetic or electric field, a device may be used in combination with a magnetic or electric plate.
IX. APPLICATIONS
a. Polynucleotide Sequencing
Generally, the methods and compositions provided herein are useful for preparation of polynucleotide fragments for downstream applications such as sequencing. Sequencing may be performed by any available technique. For example, sequencing may be performed by the classic Sanger sequencing method. Sequencing methods may also include: high-throughput sequencing, pyrosequencing, sequencing-by-synthesis, single-molecule sequencing, nanopore sequencing, sequencing-by-ligation, sequencing-by-hybridization, RNA-Seq (Illumina), Digital Gene Expression (Helicos), next generation sequencing, single molecule sequencing by synthesis (SMSS) (Helicos), massively-parallel sequencing, clonal single molecule Array (Solexa), shotgun sequencing, Maxim-Gilbert sequencing, primer walking, and any other sequencing methods known in the art.
In some cases varying numbers of fragments are sequenced. For example, in some cases about 30%-90% of the fragments are sequenced. In some cases, about 35%-85%, 40%-80%, 45%-75%, 50%-70%, 55%-65%, or 50%-60% of the fragments are sequenced. In some cases, at least about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced. In some cases less than about 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the fragments are sequenced.
In some cases sequences from fragments are assembled to provide sequence information for a contiguous region of the original target polynucleotide that is longer than the individual sequence reads. Individual sequence reads may be about 10-50, 50-100, 100-200, 200-300, 300-400, or more nucleotides in length.
The identities of the barcode tags may serve to order the sequence reads from individual fragments as well as to differentiate between haplotypes. For example, during the partitioning of individual fragments, parental polynucleotide fragments may separated into different partitions. With an increase in the number of partitions, the likelihood of a fragment from both a maternal and paternal haplotype contained in the same partition becomes negligibly small. Thus, sequence reads from fragments in the same partition may be assembled and ordered.
b. Polynucleotide Phasing
This disclosure also provides methods and compositions to prepare polynucleotide fragments in such a manner that may enable phasing or linkage information to be generated. Such information may allow for the detection of linked genetic variations in sequences, including genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) that are separated by long stretches of polynucleotides. The term “indel” refers to a mutation resulting in a colocalized insertion and deletion and a net gain or loss in nucleotides. A “microindel” is an indel that results in a net gain or loss of 1 to 50 nucleotides. These variations may exist in either a cis or trans relationship. In a cis relationship, two or more genetic variations exist in the same polynucleotide or strand. In a trans relationship, two or more genetic variations exist on multiple polynucleotide molecules or strands.
Methods provided herein may be used to determine polynucleotide phasing. For example, a polynucleotide sample (e.g., a polynucleotide that spans a given locus or loci) may be partitioned such that at most one molecule of polynucleotide is present per partition. The polynucleotide may then be fragmented, barcoded, and sequenced. The sequences may be examined for genetic variation. The detection of genetic variations in the same sequence tagged with two different bar codes may indicate that the two genetic variations are derived from two separate strands of DNA, reflecting a trans relationship. Conversely, the detection of two different genetic variations tagged with the same bar codes may indicate that the two genetic variations are from the same strand of DNA, reflecting a cis relationship.
Phase information may be important for the characterization of a polynucleotide fragment, particularly if the polynucleotide fragment is derived from a subject at risk of, having, or suspected of a having a particular disease or disorder (e.g., hereditary recessive disease such as cystic fibrosis, cancer, etc.). The information may be able to distinguish between the following possibilities: (1) two genetic variations within the same gene on the same strand of DNA and (2) two genetic variations within the same gene but located on separate strands of DNA. Possibility (1) may indicate that one copy of the gene is normal and the individual is free of the disease, while possibility (2) may indicate that the individual has or will develop the disease, particularly if the two genetic variations are damaging to the function of the gene when present within the same gene copy. Similarly, the phasing information may also be able to distinguish between the following possibilities: (1) two genetic variations, each within a different gene on the same strand of DNA and (2) two genetic variations, each within a different gene but located on separate strands of DNA.
c. Sequencing Polynucleotides from Small Numbers of Cells
Methods provided herein may also be used to prepare polynucleotides contained within cells in a manner that enables cell-specific information to be obtained. The methods enable detection of genetic variations (e.g., SNPs, mutations, indels, copy number variations, transversions, translocations, inversions, etc.) from very small samples, such as from samples comprising about 10-100 cells. In some cases, about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein. In some cases, at least about 1, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein. In other cases, at most about 5, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 cells may be used in the methods described herein.
In an example, a method comprises partitioning a cellular sample (or crude cell extract) such that at most one cell (or extract of one cell) is present per partition, lysing the cells, fragmenting the polynucleotides contained within the cells by any of the methods described herein, attaching the fragmented polynucleotides to barcodes, pooling, and sequencing.
As described elsewhere herein, the barcodes and other reagents may be contained within a partition (e.g., a capsule). These capsules may be loaded into another partition (e.g., a well) before, after, or concurrently with the loading of the cell, such that each cell is contacted with a different capsule. This technique may be used to attach a unique barcode to polynucleotides obtained from each cell. The resulting tagged polynucleotides may then be pooled and sequenced, and the barcodes may be used to trace the origin of the polynucleotides. For example, polynucleotides with identical barcodes may be determined to originate from the same cell, while polynucleotides with different barcodes may be determined to originate from different cells.
The methods described herein may be used to detect the distribution of oncogenic mutations across a population of cancerous tumor cells. For example, some tumor cells may have a mutation, or amplification, of an oncogene (e.g., HER2, BRAF, EGFR, KRAS) in both alleles (homozygous), others may have a mutation in one allele (heterozygous), and still others may have no mutation (wild-type). The methods described herein may be used to detect these differences, and also to quantify the relative numbers of homozygous, heterozygous, and wild-type cells. Such information may be used, for example, to stage a particular cancer and/or to monitor the progression of the cancer and its treatment over time.
In some examples, this disclosure provides methods of identifying mutations in two different oncogenes (e.g., KRAS and EGFR). If the same cell comprises genes with both mutations, this may indicate a more aggressive form of cancer. In contrast, if the mutations are located in two different cells, this may indicate that the cancer is more benign, or less advanced.
d. Analysis of Gene Expression
Methods of the disclosure may be applicable to processing samples for the detection of changes in gene expression. A sample may comprise a cell, mRNA, or cDNA reverse transcribed from mRNA. The sample may be a pooled sample, comprising extracts from several different cells or tissues, or a sample comprising extracts from a single cell or tissue.
Cells may be placed directly into a partition (e.g., a microwell) and lysed. After lysis, the methods of the invention may be used to fragment and barcode the polynucleotides of the cell for sequencing. Polynucleotides may also be extracted from cells prior to introducing them into a partition used in a method of the invention. Reverse transcription of mRNA may be performed in a partition described herein, or outside of such a partition. Sequencing cDNA may provide an indication of the abundance of a particular transcript in a particular cell over time, or after exposure to a particular condition.
The methods presented above provide several advantages over current polynucleotide processing methods. First, inter-operator variability is greatly reduced. Second, the methods may be carried out in microfluidic devices, which have a low cost and can be easily fabricated. Third, the controlled fragmentation of the target polynucleotides allows the user to produce polynucleotide fragments with a defined and appropriate length. This aids in partitioning the polynucleotides and also reduces the amount of sequence information loss due to the present of overly-large fragments. The methods and systems also provide a facile workflow that maintains the integrity of the processed polynucleotide. Additionally, the use of restriction enzymes enables the user to create DNA overhangs (“sticky ends”) that may be designed for compatibility with adapters and/or barcodes.
e. Partitioning of Polynucleotides, Such as Chromosomes, from Cells
In one example the methods, compositions, systems, devices, and kits provided in this disclosure may be used to partition polynucleotides, including whole chromosomes, from cells. In one example, a single cell or a plurality of cells (e.g., 2, 10, 50, 100, 1000, 10000, 25000, 50000, 100000, 500000, 1000000, or more cells) is loaded into a vessel with lysis buffer and proteinase K, and incubated for a specified period of time. Utilization of a plurality of cells will enable polynucleotide phasing, for example, by partitioning each polynucleotide to be analyzed in its own partition.
After incubation, the cell lysate is partitioned, for example by flow focusing the cell lysate into a capsule. If phasing is to be performed, flow focusing is performed such that each capsule comprises only a single analyte (e.g., a single chromosome), or only a single copy of any particular chromosome (e.g., one copy of a first chromosome and one copy of a second chromosome). In some cases, a plurality of chromosomes may be encapsulated within the same capsule, so long as the chromosomes are not the same chromosome. The encapsulation is performed under gentle flow, to minimize shearing of the polynucleotides. The capsule may be porous, to allow washing of the contents of the capsule, and introduction of reagents into the capsule, while maintaining the polynucleotides (e.g., chromosomes) within the capsules. The encapsulated polynucleotides (e.g., chromosomes) may then be processed according to any of the methods provided in this disclosure, or known in the art. The capsule shells protect the encapsulated polynucleotides (e.g., chromosomes) from shearing and further degradation. Of course, this method can also be applied to any other cellular component.
As described above, the capsule shell may be used to protect a polynucleotide from shearing. However, a capsule may also be used as a partition to enable compartmentalized shearing of a polynucleotide or other analyte. For example, in some cases a polynucleotide may be encapsulated within a capsule and then subject to ultrasonic shear, or any other suitable shearing. The capsule shell may be configured to remain intact under the shear, while the encapsulated polynucleotide may be sheared, but will remain within the capsule. In some cases, a hydrogel droplet may be used to accomplish the same end.
f. Cancer Mutation Detection and Forensics
Barcoding methods via amplification-based barcoding schemes in partitions described herein may be useful generating barcode libraries from degraded samples such as, for example, fixed formalin-fixed, paraffin-embedded (FFPE) tissue sections. Methods described herein may be capable of identifying that all amplicons within a partition originated from the same initial molecule. Indeed, with partition barcoding, information can be retained about a unique starting polynucleotide. Such identification may aid in determinations of library complexity as amplicons from different original molecules can be distinguished. Moreover, methods described herein can permit assessing unique coverage which may aid in determining variant calling sensitivity. These advantages may be particularly useful in cancer mutation detection and forensics.
g. Low Input DNA Applications (Circulating Tumor Cell (CTC) Sequencing)
Barcoding methods described herein may be useful in low polynucleotide input applications, such as, for example the sequencing of nucleic acids of circulating tumor cells (CTCs). For example, MALBAC methods described herein within a partition may aid in obtaining good data quality in low polynucleotide input applications and/or aid in filtering out amplification errors.
VIII. KITS
In some cases, this disclosure provides kits comprising reagents for the generation of partitions. The kit may comprise any suitable reagents and instructions for the generation of partitions and partitions within partitions.
In one example, a kit comprises reagents for generating capsules within droplets in an emulsion. For example, a kit may comprise reagents for generating capsules, reagents for generating an emulsion, and instructions for introducing the capsules into the droplets of the emulsion. As specified throughout this disclosure, any suitable species may be incorporated into the droplets and/or into the capsule. A kit of this disclosure may also provide any of these species, such as a polynucleotide comprising a barcode that is pre-partitioned. Similarly, as described throughout the disclosure, the capsule may be designed to release its contents into the droplets of the emulsion upon the application of a stimulus.
In another example, a kit comprises reagents for generating capsules within capsules. For example, a kit may comprise reagents for generating inner capsules, reagents for generating outer capsules, and instructions for generating capsules within capsules. As specified throughout this disclosure, any suitable species may be incorporated into the inner and/or outer capsules. A kit of this disclosure may also provide any of these species, such as a polynucleotide comprising a barcode that is pre-partitioned. Similarly, as described throughout the disclosure, the inner capsule may be designed to release its contents into the outer capsule upon the application of a stimulus.
IX. DEVICES
In some cases, this disclosure provides devices comprising partitions for the processing of analytes. A device may be a microwell array, or a microspot array, as described elsewhere in this disclosure. A device may formed in a manner that it comprises any suitable partition. In some cases, a device comprises a plurality of wells, or a plurality of spots. Of course, any partition in a device may also hold other partitions, such as a capsule, a droplet in an emulsion, and the like.
A device may be formed from any suitable material. In some examples, a device is formed from a material selected from the group consisting of fused silica, soda lime glass, borosilicate glass, poly(methyl methacrylate), sapphire, silicon, germanium, cyclic olefin copolymer, polyethylene, polypropylene, polyacrylate, polycarbonate, plastic, and combinations thereof.
In some cases, a device comprises channels for the flow of fluids into and between partitions. Any suitable channels may be used. A device may comprise a fluid inlet and a fluid outlet. The inlet and outlet may be attached to liquid handling devices to introduce species into the device. The device may be sealed, before or after introduction of any species.
Materials that are hydrophilic and/or hydrophobic may be used in different parts of the device. For example, in some cases a device of this disclosure comprises a partition with an interior surface comprising a hydrophilic material. In some cases a surface exterior to the partitions comprises a hydrophobic material. In some cases, a fluid flow path is coated with a hydrophobic or hydrophilic material.
As will be appreciated, the instant disclosure provides for the use of any of the compositions, libraries, methods, devices, and kits described herein for a particular use or purpose, including the various applications, uses, and purposes described herein. For example, the disclosure provides for the use of the compositions, methods, libraries, devices, and kits described herein in partitioning species, in partitioning oligonucleotides, in stimulus-selective release of species from partitions, in performing reactions (e.g., ligation and amplification reactions) in partitions, in performing nucleic acid synthesis reactions, in barcoding nucleic acid, in preparing polynucleotides for sequencing, in sequencing polynucleotides, in polynucleotide phasing, in sequencing polynucleotides from small numbers of cells, in analyzing gene expression, in partitioning polynucleotides from cells, in mutation detection, in neurologic disorder diagnostics, in diabetes diagnostics, in fetal aneuploidy diagnostics, in cancer mutation detection and forensics, in disease detection, in medical diagnostics, in low input nucleic acid applications, such as circulating tumor cell (CTC) sequencing, in a combination thereof, and in any other application, method, process or use described herein.
EXAMPLES Example 1 Production of a Library of Forked Adapters Comprising Barcode Sequences by Asymmetric PCR and Addition of a Partially Complementary Universal Sequence
This example provides methods for the manufacture of forked adapters comprising barcode sequences compatible with next generation sequencing technologies (e.g., ILLUMINA). In this example, the barcode is placed in position 207 as depicted in FIG. 2.
With reference to FIG. 4, a single-stranded adapter-barcode polynucleotide sequence 401 comprising a first immobilization region 402, a barcode region 403, and a first sequencing primer region 404 is synthesized. The barcode region 403 is a seven nucleotide random sequence synthesized by including equimolar concentrations of A, G, T, and C in each coupling step.
Following synthesis, the single-stranded adapter-barcode polynucleotide 401 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 0.1 polynucleotides. The droplets also comprise reagents for amplification of the single-stranded adapter-barcode polynucleotide 401, by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts) and a DNA intercalating dye (e.g., ethidium bromide). The reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification. The polynucleotides are amplified and the reaction proceeds through an exponential phase of amplification 410, which produces double-stranded products 405, and a linear phase amplification 411, which produces single-stranded products 406.
The droplets are sorted on a fluorescence assisted cell sorter (FACS) 412 to collect droplets comprising amplified polynucleotides. A partially complementary universal sequence 407 is added to the partitions to generate a partially annealed fork structure 413. Partially complementary universal sequence 407 comprises a second immobilization region 408 and a second sequencing primer region 409, the latter of which comprises a T overhang that is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
Example 2 Fragmentation and Barcoding with Fragmentase
A single-stranded adapter-barcode polynucleotide sequence (e.g., FIG. 4: 401) comprising a first immobilization region 402, a barcode region 403, and a first sequencing primer region 404 is synthesized, partitioned, amplified, and sorted as described in Example 1, or by any other method described in this disclosure. Interfacial polymerization is performed on the droplet comprising the single-stranded adapter-barcode polynucleotide sequence, to generate a plurality of capsules comprising a library of single-stranded adapter-barcode polynucleotide sequences 406, where each (or most) sequences in the library differ in the sequence of their respective barcode regions 403. Thus, a library of encapsulated single-stranded adapter-barcode polynucleotides is generated.
Two mixtures are prepared. Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded), a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE), and a partially complementary universal sequence (e.g., FIG. 4: 407). A second mixture Z2 comprises the library of encapsulated single-stranded adapter-barcode polynucleotides, generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme. Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
Mixtures Z1 and Z2 are combined and a capsule within a capsule is formed according to methods described elsewhere in this disclosure, such as flow focusing. FIG. 5 illustrates a capsule within a capsule produced according to the method described above. The outer capsule 501 comprises an inner capsule 502 and medium 504. The inner capsule 502 is one member of the library of encapsulated single-stranded adapter-barcode polynucleotides. Thus, inner capsule 502 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 503, which can be used to attach the same barcode to a polynucleotide within a partition, such as outer capsule 501.
The medium 504 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 504 comprises target polynucleotide 505, the partially complementary universal sequence 506, and the enzyme mix 507 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture inner capsule 502, releasing its contents into outer capsule 501. The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide. The single-stranded adapter-barcode polynucleotide 503 hybridizes with the partially complementary universal sequence 506, forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide. The thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide. The outer capsule 501 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced. Additional preparation steps (e.g., bulk amplification, size selection, etc.) may then be performed as needed prior to sequencing.
In some cases, mixture Z1 comprises multiple versions of the partially complementary universal sequence 506, where each version has its own sample-specific barcode.
Moreover, although the example described above utilizes a thermally stable ligase to attach the forked adapter comprising the barcode sequence to the target polynucleotide, PCR can also be used to accomplish this step, as described elsewhere in this disclosure.
Example 3 Fragmentation and Barcoding by Sonication
A library of encapsulated single-stranded adapter-barcode polynucleotides is generated as described in Example 2, or by any other suitable method described in this disclosure. Target polynucleotides (i.e., polynucleotides to be fragmented) are partitioned into capsules. The capsules comprising the target polynucleotides are configured to withstand ultrasonic stress. The capsules comprising the target polynucleotides are exposed to ultrasonic stress (e.g., COVARIS Focused-Ultrasonicator) and the target polynucleotide is fragmented, generating fragmented target polynucleotide capsules.
A mixture Z1 is prepared, comprising the library of encapsulated single-stranded adapter-barcode polynucleotides (e.g., FIG. 4: 406), the fragmented target polynucleotide capsules, a partially complementary universal sequence (e.g., FIG. 4: 407), an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase), and appropriate buffers. A capsule within capsule is generated according to the method described elsewhere in this disclosure, such as flow focusing.
FIG. 6 illustrates capsules within a capsule produced according to the methods described above. The outer capsule 601 comprises a plurality of inner capsules 602 and 605 and medium 604. The inner capsules 602 and 605 include capsules comprising single-stranded adapter-barcode polynucleotides 603 and capsules comprising fragmented target polynucleotide 606, respectively. Inner capsule 602 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 603, which can be used to attach the same barcode to a polynucleotide within a partition, such as the fragmented polynucleotides 606 contained within inner capsules 605.
The medium 604 contains the contents of mixture Z1, described above. More specifically, medium 604 comprises a partially complementary universal sequence 607, an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 608, and appropriate buffers.
Inner capsules 605 comprising fragmented target polynucleotides 606 are exposed to a stimulus to rupture them and release their contents into the contents of outer capsule 601. The T4 polymerase blunts the ends of the fragmented target polynucleotides; the Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide. The T4 polymerase and Taq polymerase is then heat-inactivated and a stimulus is applied to release the contents of inner capsule 602 into outer capsule 601. The single-stranded adapter-barcode polynucleotide 603 hybridizes with the partially complementary universal sequence 607, forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide. The thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating a barcoded target polynucleotide. The outer capsule 601 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
As described in Example 2, in some cases Z1 can comprise multiple versions of the partially complementary universal sequence 607. Furthermore, although this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
Example 4 Generation of Forked Adapters by Single Primer Isothermal Amplification (SPIA) and Restriction Digestion
This example demonstrates the synthesis of a forked adapter by SPIA and restriction digestion. FIG. 7 provides an example of a product (or intermediate) that may be generated according to the methods of this example. With reference to FIG. 7, a hairpin adapter 701 (SEQ ID NO: 2) is shown that can be used as a precursor to a forked-adapter as described elsewhere in this disclosure. In this example, the hairpin adapter is synthesized as a single-stranded amplification product utilizing SPIA. The hairpin adapter 701 comprises a double-stranded region 702, a 3′-T overhang for AT ligation 703, and a region that can be cut by a restriction enzyme 704 (i.e., between positions 33 and 34). The hairpin adapter may comprise a barcode region and functional regions, such as immobilization regions and regions for annealing of sequencing primers.
Cutting of the adapter (e.g., between positions 33 and 34) generates the forked-adapter depicted in FIG. 8a (SEQ ID NOs: 3-4). The adapter is cut by introducing an oligonucleotide sequence complementary to the region to be cut and exposing the annealed adapter to a restriction enzyme. Ligation of the forked-adapter region depicted in FIG. 8a to a target polynucleotide results in the structure depicted in FIG. 8b (SEQ ID NOs: 5-6). With reference to FIG. 8, the underlined portions of the sequences in FIG. 8b comprise the target polynucleotide with 3′-A overhangs compatible for ligation with the forked adapter depicted in FIG. 8a . The sequences shown in FIG. 8b (SEQ ID NOs: 5-6) are then amplified by polymerase chain reaction to produce SEQ ID NO: 7 (amplification product of SEQ ID NO: 5) and SEQ ID NO: 8 (amplification product of SEQ ID NO: 6), shown in FIG. 8c . In FIG. 8c , SEQ ID NO: 7 represents an amplification product of SEQ ID NO: 5 that adds a first immobilization sequence (underlined 5′ portion) and a second immobilization sequence (underlined 3′ portion) to SEQ ID NO: 5. SEQ ID NO: 8 represents an amplification product of SEQ ID NO: 6 that replaces the unhybridized portions of SEQ ID NO: 6 with different sequences (underlined 3′ portion and underlined 5′ portion). Additionally, SEQ ID NO: 8 includes a six nucleotide barcode (TAGTGC; bolded) within the 5′ unhybridized region of the polynucleotide. The amplification product therefore comprises barcoded target polynucleotide sequence (represented by 111), immobilization sequences, and a barcode.
Example 5 Additional Forked Adapters by Single Primer Isothermal Amplification (SPIA) and Restriction Digestion
This example demonstrates the synthesis of a forked adapter as depicted in FIG. 9a (SEQ ID NOs: 9-10) by SPIA and restriction digestion, where N represents A, T, G, or C. FIG. 9b shows the forked-adapter in single-stranded format (SEQ ID NO: 11), where the single stranded format is capable of forming a hairpin structure. Cutting the hairpin structure at the position designated by the asterisk yields the forked adapter shown in FIG. 9 a.
The template for the SPIA will be the sequence shown in FIG. 9c (SEQ ID NO: 12). In FIG. 9c , “R” represents a region of RNA. FIG. 9d shows the hairpin structure formed by the sequence in FIG. 9c . The sequence in FIG. 9d (SEQ ID NO: 12) is treated with polymerase to add nucleotides to the 3′ end, generating the sequence shown in FIG. 9e (SEQ ID NO: 13). The sequence in FIG. 9e (SEQ ID NO: 13) is then treated with RNase H, which degrades RNA hybridized to DNA, yielding the sequence in FIG. 9f (SEQ ID NO: 14).
Strand displacement SPIA is then performed on SEQ ID NO: 14. The primer in the strand displacement amplification is of the form RRRRRRRRRRRRR (i.e., R13). This primer is an RNA primer that is one base longer than the unhybridized 3′ terminus of SEQ ID NO: 14 (i.e., N12) (FIG. 9f ). More specifically, as shown in FIG. 9f , the 3′ terminus of SEQ ID NO: 14 contains twelve N nucleotides. The RNA primer contains 13 nucleotides. Nucleotides 2-13 of the RNA primer are complementary with the twelve unhybridized N nucleotides of SEQ ID NO: 14. Nucleotide 1 of the RNA primer is complementary with the first hybridized base (going from 3′ to 5′), in this case, T. The RNA primer displaces the A and generates the double-stranded extension product shown in FIG. 9g (SEQ ID NOs: 15-16). Because only one primer is present, the reaction produces multiple copies of the single-stranded product. The single-stranded amplification products are treated with RNase H to generate the single-stranded amplification products shown in FIG. 9h (SEQ ID NO: 17). FIG. 9i shows this sequence in 5′-3′ format (SEQ ID NO: 17). FIG. 9j shows this sequence in hairpin format (SEQ ID NO 17).
The hairpin adapter shown in FIG. 9j is then ligated to a fragmented polynucleotide with a 3′-A overhang. The hairpin is cleaved between the A and C residues separated by the curved line in FIG. 9j by adding an oligonucleotide complementary to that region and cutting with a restriction enzyme. This generates a forked adapter. PCR amplification is then conducted, as described in Example 4, to add immobilization regions and barcodes to the forked adapter that is attached to the target polynucleotide.
Example 6 Generation of Forked Adapters Comprising Barcodes by Exponential PCR and Hybridization
This example demonstrates the production of forked adapters comprising barcodes by hybridization. FIG. 10a shows the exemplary forked adapter provided in FIG. 8a . As described in Example 4, this adapter may be ligated to a target polynucleotide and then an amplification reaction may be performed to add additional functional sequences, including a barcode. However, a barcode (and other functional sequences) may also be incorporated directly into the forked adapter, prior to attachment of the forked adapter to the target polynucleotide. For example, FIG. 10b shows the forked adapter of FIG. 10a , with the addition of a first immobilization region (underlined) and a seven nucleotide barcode region (bold/underline; “N”).
The barcoded forked adapter of FIG. 10b is produced by first synthesizing SEQ ID NO: 18 as a single strand. The diversity in the barcode region is generated using an equimolar mixture of A, G, T, and C, as described throughout this disclosure. Droplet-based PCR is performed, as described in Example 1. However, one DNA primer and one RNA primer are used to amplify SEQ ID NO: 18 in the droplets. The amplification is conducted in the presence of an intercalating dye, and droplets comprising amplified SEQ ID NO: 18 are isolated, as described in Example 1. FIG. 10c shows the double-stranded amplification product. The underlined portion of SEQ ID NO: 19 is an RNA strand derived from the RNA primer. The sequences shown in FIG. 10c are then treated with RNase H, which digests the underlined RNA region, yielding the construct shown in FIG. 10d . In order to generate a forked construct, a partially complementary universal sequence (SEQ ID NO: 21) is added to the construct shown in FIG. 10d , producing the product shown in FIG. 10e . The advantage to utilizing this process is that it utilizes the significantly greater amplification of polynucleotides provided by exponential PCR versus the linear amplification of polynucleotides provided by SPIA.
Example 7 Dual Indexing Approach
This example demonstrates an approach for synthesis of barcodes for dual-index reads. A dual-index read is a read of both strands of a double-stranded fragment, using barcodes attached to each strand. FIG. 11 shows an example of the synthesis of barcodes for a dual-indexing approach and an example use of the barcodes in a capsules in a capsule configuration.
As shown in FIG. 11a , a first single-stranded adapter-barcode polynucleotide sequence 1101 comprising a first immobilization region 1102, a first barcode region 1103, and a first sequencing primer region 1104 is synthesized. In parallel, as shown in FIG. 11b , a second single-stranded adapter-barcode polynucleotide sequence 1131, comprising a second immobilization region 1132, a second barcode region 1133, and a second sequencing primer region 1134 is synthesized. In some cases, barcode regions 1103 and 1133 are of the same sequence. In other cases, barcode regions 1103 and 1133 are of different sequences or of partially different sequences.
Following synthesis, the single-stranded adapter-barcode polynucleotides 1101 (FIG. 11a ) and 1131 (FIG. 11b ) are, in parallel, diluted into aqueous droplets in a water-in-oil emulsion. The droplets also comprise reagents for amplification of the single-stranded adapter-barcodes polynucleotide 1101 (FIG. 11a ) and 1131 (FIG. 11b ) respectively, by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts) and a DNA intercalating dye (e.g., ethidium bromide). The reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification. The polynucleotides 1101 (FIG. 11a ) and 1131 (FIG. 11b ) are amplified and the reaction proceeds through an exponential phase of amplification 1110, which produces double-stranded products 1105 (FIG. 11a ) and 1135 (FIG. 11b ), and a linear phase amplification 1111, which produces single-stranded products 1106 (FIG. 11a ) and 1136 (FIG. 11b ) respectively.
The droplets are sorted on a fluorescence assisted cell sorter (FACS) 1112 to collect droplets comprising amplified polynucleotides.
Interfacial polymerization is then performed on the droplets comprising the single-stranded adapter- barcode polynucleotide sequences 1106 and 1136 droplets respectively, to generate two types of capsules 1120 (FIG. 11a ) and 1150 (FIG. 11b ), each comprising one of single-stranded adapter barcode polynucleotide sequences 1106 or 1136 respectively.
Two mixtures are prepared. Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded) 1170 and a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE). A second mixture Z2 comprises capsules 1120 and 1180, generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme. Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
Mixtures Z1 and Z2 are combined and a capsule within a capsule is formed according to methods described elsewhere in this disclosure, such as flow focusing. FIG. 11c illustrates capsules within a capsule produced according to the method described above. The outer capsule 1160 comprises capsules 1120 and 1150 and medium 1190. Thus, capsules 1120 and 1150 each comprise multiple copies of single-stranded adapter- barcode polynucleotides 1106 and 1136, respectively, and can be used to attach barcodes 1103 and 1133 to a polynucleotide within a partition, such as target polynucleotide 1170 in medium 1190 of outer capsule 1160.
The medium 1190 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 1190 comprises target polynucleotide 1170 and the enzyme mix 1180 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
Upon generation of the capsules within a capsule, and exposure of the capsules within the capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture capsules 1120 and 1150, releasing their contents into medium 1190 of outer capsule 1160. The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide. The single-stranded adapter-barcode polynucleotide 1106 hybridizes with single-stranded adapter-barcode polynucleotide 1136, forming a forked adapter, comprising barcode regions 1103 and 1133, with a 3′-T overhang that is compatible with the 3′-A overhang (not shown) on the fragmented target polynucleotide. The thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide. The outer capsule 1160 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
Moreover, although the example described above utilizes a thermally stable ligase to attach the forked adapter comprising the barcode sequence to the target polynucleotide, PCR can also be used to accomplish this step, as described elsewhere in this disclosure.
Example 8 Production of a Forked Adapter Comprising Barcode Sequences by Bead Emulsion PCR and Addition of a Partially Complementary Universal Sequence
As shown in FIG. 14a , a single-stranded adapter-barcode sequence 1401 comprising a first immobilization region 1402, a barcode region 1403, and a first sequencing primer region 1404 is synthesized. Following synthesis, the single-stranded adapter-barcode sequence 1401 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 1 polynucleotide. The droplets also comprise first beads 1405 that are linked, via a photolabile linker, to one or more copies of an RNA primer 1406 complementary to a sequence comprised in the first sequencing primer region 1404; a DNA primer complementary to a sequence (not shown) comprised in the first immobilization region 1402; and reagents necessary for amplification (e.g., polymerase, dNTPs, buffer, salts) of single-stranded adapter-barcode sequence 1401. The polynucleotides are amplified 1407 which produces double-stranded products 1408 both attached to the first beads 1405 to form structure 1420 and in solution (not shown).
The emulsion is then broken and the emulsion components are pooled to form a product mixture. As shown in FIG. 14b , the liberated beads are then washed 1409 (via centrifugation) several times with appropriate medium, treated with sodium hydroxide (NaOH) 1410 to denature the double-stranded products attached to the first beads 1405, and then further washed 1411. After denaturation 1410 and washing 1411 of structure 1420, the resulting structure 1430 comprises a single-stranded complement 1412 to the single-stranded adapter-barcode sequence 1401, comprising a complementary immobilization region 1413, a complementary barcode region 1414, and a complementary sequencing primer region 1415. As shown, the complementary sequencing primer region 1415 comprises the RNA primer 1406. Structure 1430 is then resuspended in an appropriate medium.
Next, as shown in FIG. 14c , second beads 1416 that comprise one or more copies of a DNA polynucleotide 1417 complementary to the complementary immobilization region 1413 are then added to the medium. Via the complementary DNA polynucleotide 1417 and the complementary immobilization region 1413 of the single-stranded complement 1412, the second beads 1416 bind to the single-stranded complement 1412. The single stranded complement is now bound at one end to first bead 1405 and at its other end second bead 1416 to form structure 1440.
As shown in FIG. 14d , structure 1440 is then centrifuged 1418 using a glycerol gradient to separate structure 1440 from structure 1430 not comprised in structure 1440. In cases where the second beads 1416 are magnetic, a magnetic separation may be used as an alternative. The product is then treated with NaOH 1419 to denature the single-stranded complement 1412 from the second bead 1416, resulting in regeneration of structure 1430. Structure 1430 is then subject to several rounds of washing (via centrifugation) to remove second beads 1416. Single-stranded complement 1412, attached to structure 1430, represents a single-stranded barcode adapter.
As shown in FIG. 14e , single-stranded complement 1412 can be used to generate a forked adapter. To generate a forked adapter 1450, the single-stranded complement 1412 is then released 1424 from structure 1430 with light and then combined 1425 with a universal complementary sequence 1426 or is first combined 1425 with a universal complementary sequence 1426 and then released 1424 from structure 1430. In order to generate ligatable ends, RNAase H is used to digest the RNA primer 1406 of the single-stranded complement 1412 and a Type IIs restriction enzyme is used to generate a single base T overhang on the universal complementary sequence 1426. The T overhang is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
Example 9 Production of a Forked Adapter Comprising Barcode Sequences by Bead Emulsion PCR and Addition of a Partially Complementary Universal Sequence
As shown in FIG. 15a , a single-stranded adapter-barcode sequence 1501 comprising a first immobilization region 1502, a barcode region 1503, and a first sequencing primer region 1504 is synthesized. Following synthesis, the single-stranded adapter-barcode sequence 1501 is diluted into aqueous droplets in a water-in-oil emulsion such that each droplet comprises, on average, 1 polynucleotides. The droplets also comprise first beads 1505 that are linked, via a photolabile linker, to one or more copies of an RNA primer 1506 complementary to a sequence comprised in the first immobilization region 1502; a DNA primer complementary to a sequence (not shown) comprised in the first sequencing primer region 1502; and reagents necessary for amplification (e.g., polymerase, dNTPs, buffer, salts) of single-stranded adapter-barcode sequence 1501. The polynucleotides are amplified 1507 which produces double-stranded products 1508 both attached to the first beads 1505 to form structure 1520 and in solution (not shown).
The emulsion is then broken and the emulsion components are pooled to form a product mixture. As shown in FIG. 15b , the liberated beads are then washed 1509 (via centrifugation) several times with appropriate medium, treated with sodium hydroxide (NaOH) 1510 to denature the double-stranded products attached to the first beads 1505, and then further washed 1511. After denaturation 1510 and washing 1511 of structure 1520, the resulting structure 1430 comprises a single-stranded complement 1512 to the single-stranded adapter-barcode sequence 1501, comprising a complementary immobilization region 1513, a complementary barcode region 1514, and a complementary sequencing primer region 1515. As shown, the complementary sequencing primer region 1515 comprises the RNA primer 1506. Structure 1530 is then resuspended in an appropriate medium.
Next, as shown in FIG. 15c , second beads 1516 that comprise one or more copies of a DNA polynucleotide 1517 complementary to the complementary sequencing primer region 1515 are then added to the medium. Via the complementary DNA polynucleotide 1517 and the complementary sequencing primer region 1515 of the single-stranded complement 1512, the second beads 1416 bind to the single-stranded complement 1512. The single stranded complement is now bound at one end to first bead 1505 and at its other end second bead 1516 to form structure 1540.
As shown in FIG. 15d , structure 1540 is then centrifuged 1518 using a glycerol gradient to separate structure 1540 from structure 1530 not comprised in structure 1540. In cases where the second beads 1516 are magnetic, a magnetic separation may be used as an alternative. The product is then treated with NaOH 1519 to denature the single-stranded complement 1512 from the second bead 1516, resulting in regeneration of structure 1530. Structure 1530 is then subject to several rounds of washing (via centrifugation) to remove second beads 1516. Single-stranded complement 1512, attached to structure 1530, represents a single-stranded barcode adapter.
As shown in FIG. 15e , single-stranded complement 1512 can be used to generate a forked adapter. To generate a forked adapter 1550, the single-stranded complement 1512 is then optionally released 1524 from structure 1530 with light and then combined 1525 with a universal complementary sequence 1526. In order to generate ligatable ends, a Type IIs restriction enzyme is used to generate a single base T overhang on the universal complementary sequence 1526. The T overhang is compatible with the A overhang on a polynucleotide target to be sequenced (not shown).
Example 10 Production of a Forked Adapter Template Barcode Sequences by Bead Emulsion PCR and an Adapter Derived Therefrom
As shown in FIG. 16, structure 1600 comprising a magnetic bead (1601)-bound single-stranded adapter-barcode sequence 1602 is produced according to methods described in Example 8, Example 9, or any other method described herein. Next, structure 1600 is partitioned into capsules (or alternatively, another emulsion) 1620 by methods described herein, for example, interfacial polymerization. The capsules 1620 also comprise reagents for amplification of the single-stranded adapter-barcode sequence 1602, by asymmetric PCR (e.g., polymerase, primers, dNTPs, buffer, salts). The reverse primer is present in excess of the forward primer, or vice versa, enabling asymmetric amplification. Single-stranded adapter-barcode sequence 1602 is amplified 1603 and the reaction proceeds through a linear phase amplification 1604, which produces single-stranded adapter product 1605, complementary to single-stranded barcode adapter-template 1602. At this juncture, capsules 1620 comprise both single-stranded adapter 1605 in solution and magnetic bead (1601)-bound single-stranded adapter-barcode sequence 1602. Capsules 1620 are then separated from those not comprising beads (and thus templates 1602 and single-stranded adapters 1605) by magnetic separation 1606. Capsules 1620 may be ruptured and forked adapters generated as described in Example 9.
Example 11 Barcoding with Bead Emulsion PCR and Fragmentation with Fragmentase
As shown in FIG. 17, structure 1700 comprising a magnetic bead (1701)-bound single-stranded adapter-barcode sequence 1702 is produced according to methods described in Example 8, Example 9, or any other method described herein. Interfacial polymerization is performed on the droplet comprising the structure 1700, to generate a capsule 1704 comprising single-stranded adapter-barcode sequence 1702 attached, via a photolabile linker, to a bead 1701.
Two mixtures are prepared. Mixture Z1 comprises a target polynucleotide (i.e., a polynucleotide to be fragmented and barcoded), a fragmentase enzyme (e.g., NEBNEXT DSDNA FRAGMENTASE), and a partially complementary universal sequence. A second mixture Z2 comprises capsule 1704 generated as described above and magnesium chloride in a concentration sufficient to activate the fragmentase enzyme. Mixture Z1, Z2, or both Z1 and Z2 also comprise T4 polymerase, Taq polymerase, and a thermostable ligase.
Mixtures Z1 and Z2 are combined and a capsule within a capsule is formed according to methods described elsewhere in this disclosure, such as flow focusing. FIG. 17 illustrates a capsule within a capsule produced according to the method described above. The outer capsule 1703 comprises an inner capsule 1704 and medium 1705. The inner capsule 1704 is one member of a library of encapsulated, bead-bound single-stranded barcode adapters. Thus, inner capsule 1704 comprises multiple copies of structure 1700, which can be used to generate a free single-stranded adapter-barcode sequence 1702 and attach the same barcode adapter to a polynucleotide within a partition, such as outer capsule 1703.
The medium 1705 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 1705 comprises target polynucleotide 1706, the partially complementary universal sequence 1707, and the enzyme mix 1708 comprising fragmentase, T4 polymerase, Taq polymerase, thermostable ligase, magnesium chloride, and appropriate buffers.
Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the enzymes process the target polynucleotide. More specifically, the fragmentase fragments the target polynucleotide and the T4 polymerase blunts the ends of the fragmented target polynucleotide. The fragmentase and T4 polymerase are then heat inactivated and a stimulus is used to rupture inner capsule 1704, releasing its contents into outer capsule 1703. The Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide. The single-stranded adapter-barcode sequence 1702 hybridizes with the partially complementary universal sequence 1707 and is released from the bead with light, forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide. The thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating barcoded target polynucleotide. The outer capsule 1703 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced. Additional preparation steps (e.g., bulk amplification, size selection, etc.) may be performed as needed prior to sequencing.
In some cases Z1 can comprise multiple versions of the partially complementary universal sequence 1707. Furthermore, although this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
Example 12 Barcoding with Bead Emulsion PCR and Fragmentation by Sonication
As shown in FIG. 18, structure 1800 comprising a magnetic bead (1801)-bound single-stranded adapter-barcode sequence 1802 is produced according to methods described in Example 8, Example 9, or any other method described herein. Interfacial polymerization is performed on the droplet comprising the structure 1800, to generate a capsule 1803 comprising single-stranded adapter-barcode sequence 1802 attached, via a photolabile linker, to a bead 1801. Target polynucleotides (i.e., polynucleotides to be fragmented) are partitioned into capsules 1804. The capsules 1804 comprising the target polynucleotides are configured to withstand ultrasonic stress. The capsules 1804 comprising the target polynucleotides are exposed to ultrasonic stress (e.g., COVARIS Focused-Ultrasonicator) and the target polynucleotide is fragmented, generating fragmented target polynucleotide capsules.
A mixture Z1 is prepared, comprising capsule 1803, the fragmented target polynucleotide capsules 1804, a partially complementary universal sequence 1805, an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 1806, and appropriate buffers. A capsule within capsule is generated according to the method described elsewhere in this disclosure, such as flow focusing.
FIG. 18 illustrates capsules within a capsule produced according to the methods described above. The outer capsule 1807 comprises capsules 1803 and 1804 and medium 1808. The inner capsules 1803 and 1804 include capsules comprising structure 1800 and capsules comprising fragmented target polynucleotide 1809, respectively. Inner capsule 1803 comprises multiple copies of structure 1800, which can be used to generate a free single-stranded barcode adapter 1802 and attach the same barcode adapter to a polynucleotide within a partition, such as the fragmented polynucleotides 1809 contained within inner capsules 1804.
The medium 1808 contains the contents of mixture Z1, described above. More specifically, medium 1808 comprises a partially complementary universal sequence 1805, an enzyme mixture (T4 polymerase, Taq polymerase, and a thermostable ligase) 1806, and appropriate buffers.
Inner capsules 1804 comprising fragmented target polynucleotides 1809 are exposed to a stimulus to rupture them and release their contents into the contents of outer capsule 1807. The T4 polymerase blunts the ends of the fragmented target polynucleotides; the Taq polymerase adds 3′-A overhangs to the fragmented, blunt-ended target polynucleotide. The T4 polymerase and Taq polymerase is then heat-inactivated and a stimulus is applied to release the contents of inner capsule 1803 into outer capsule 1807. The single-stranded adapter-barcode sequence 1802 hybridizes with the partially complementary universal sequence 1805 and the adapter is released from the bead with light forming a forked adapter with a 3′-T overhang that is compatible with the 3′-A overhang on the fragmented target polynucleotide. The thermostable ligase ligates the forked adapter to the fragmented target polynucleotide, generating a barcoded target polynucleotide. The outer capsule 1807 is then ruptured, samples from all outer capsules are pooled, and the target polynucleotides are sequenced.
In some cases Z1 can comprise multiple versions of the partially complementary universal sequence 1807. Furthermore, although this example demonstrates barcoding of a target polynucleotide by utilizing a thermostable ligase, PCR can also be used to accomplish this step.
Example 13 Barcoding with Multiple Annealing and Looping-Based Amplification (MALBAC)
As shown in FIG. 19a , a primer comprising SEQ ID NO: 36 is prepared. The primer comprises a barcode region (designated “Barcode”), a primer sequencing region (designated “PrimingSeq”), and a eight-nucleotide variable region (designated as “ ”) that may comprise any combination of A, T, C, or G. The primer shown in FIG. 19 is combined with a target polynucleotide (indicated by the loop in FIG. 19), along with a polymerase (e.g., Vent, exo+DeepVent, exo−DeepVent) possessing of strand-displacement activity into a partition (e.g., a capsule, droplet of an emulsion, etc.). In some cases, a non strand-displacing polymerase (e.g., Taq, PfuUltra) is used. The partition is then subject to MALBAC amplification. Appropriate MALBAC cycling conditions are known and are, described for example, in Zong et al., Science, 338(6114), 1622-1626 (2012), which is incorporated herein by reference, in its entirety.
A looped MALBAC product is produced as shown in FIG. 19b as SEQ ID NO: 23. The looped MALBAC product comprises the original primer shown in FIG. 19a , the target polynucleotide to be barcoded oriented in a loop, and a region complementary to and hybridized to the original primer sequence. The partition is broken and the contents recovered. In some cases, a plurality of partitions are generated. The partitions are collectively broken, the contents of each recovered, and then pooled.
Next, the generated MALBAC product shown in FIG. 19b is treated with a restriction enzyme (e.g., BfuC1 or similar) to generate a 4-basepair overhang (in this case, GATC shown in italics) on the MALBAC product. This structure is represented by SEQ ID NO: 24 and shown in FIG. 19c . A forked adapter, shown in FIG. 19d as SEQ ID NO: 25 and SEQ ID NO: 37, comprising an overhang (in this case, CTAG shown in bold) complementary to the overhang generated on the MALBAC product. The forked adapter is mixed with the MALBAC product in FIG. 19c and the complementary regions hybridize. A thermostable ligase is used to ligate the forked adapter and MALBAC product together to form the desired structure FIG. 19e as SEQ ID NO: 26. Additional amplification methods (e.g., PCR) can be used to add additional regions (e.g., immobilization regions, additional barcodes, etc.) to the forked adapter.
In some cases, other basepair overhangs (e.g., 1 basepair overhang-10 basepair overhang) may be desired. Restriction enzymes used to generate these overhangs may be used as an alternative, including those described herein, where desired. In one example, a two basepair overhang is generated on the MALBAC product using TaqαI.
As an alternative, the primer shown in FIG. 19a can be designed such that an RNA primer sequence is placed 5′ of the barcode region, such that an RNAase is used to generate an overhang. As shown in FIG. 19f , MALBAC product 1900 comprises an RNA primer sequence 1901 placed 5′ of a barcode region 1902. MALBAC product 1900 also comprises a sequencing primer region 1903, the target polynucleotide 1904, a complementary sequencing primer region 1905, a complementary barcode region 1906, and a region 1907 complementary to the RNA primer sequence 1901. MALBAC product 1900 is treated with an RNAse H 1908 and the RNA primer region sequence 1901 is digested to yield a 2-6 basepair overhang 1909 on MALBAC product 1900 to give structure 1920. A universal complementary region 1910 is then added to structure 1910 that comprises a region complementary to the overhang on structure 1910. Universal complementary region 1910 then hybridizes with structure 1920 and a thermostable ligase is used to ligate universal complementary region 1910 to structure 1920.
Example 14 Barcoding with Multiple Annealing and Looping-Based Amplification (MALBAC)
As shown in FIG. 20, a template 2000 comprising a barcode region is combined with agents 2001 necessary for PCR into a capsule 2002, using, for example interfacial polymerization or any other method described herein. PCR is used to generate a MALBAC primer from the template 2000. Next, the capsule 2000 is encapsulated into an outer capsule 2003 that also comprises a mixture 2004 that comprises a target polynucleotide 2005 to be barcoded and reagents 2006 necessary for MALBAC amplification (e.g., DeepVent polymerase, dNTPs, buffer). Capsule 2002 is broken upon proper exposure of capsule 2002 to a stimulus designed to rupture capsule 2002, the contents of capsule 2002 mix with those of mixture 2004. MALBAC amplification of the target polynucleotide 2005 commences to produce a MALBAC product similar to that described as that shown as 1900 in FIG. 19 f.
The outer capsule 2003 is then broken with the appropriate stimulus and the contents recovered. The MALBAC product is then treated with an appropriate restriction enzyme and coupled to a forked adapter in a matter as described in Example 13. Additional downstream preparation steps (e.g., bulk amplification, size selection, etc.) are then performed as needed.
Example 15 Barcoding with Multiple Annealing and Looping-Based Amplification (MALBAC)
As shown in FIG. 21a , a MALBAC primer 2100 is prepared. MALBAC primer 2100 comprises a sequence priming region 2101 and an 8-nucleotide variable region 1902. Primer 2100 is combined with target polynucleotide 2103, along with a polymerase (e.g., Vent, exo+DeepVent, exo−DeepVent) possessing of strand-displacement activity into a partition (e.g., a capsule, emulsion, etc.). In some cases, a non strand-displacing polymerase (e.g., Taq, PfuUltra) is used. The partition is then subject to MALBAC amplification 2104.
A looped MALBAC product 2110 is produced and comprises sequencing priming region 2101, target polynucleotide 2103, and a complementary sequence priming region 2105. Shown in linear form 2120 in FIG. 21b , MALBAC product 2110 is then contacted with another primer 2130 that comprises a sequencing primer region 2106, a barcode region 2107, and an immobilization region 2108. Primer 2130 is produced using asymmetric digital PCR. Using a single cycle of PCR, the primer is used to generate double-stranded product 2140 that comprises primer 2130, and, thus, barcode region 2107.
Double-stranded product 2140 may be then denatured and subsequently contacted with another primer 2150 shown in FIG. 21c . Primer 2150 comprises a barcode region 2109, a sequencing primer region 2111, and an immobilization region 2112. In the presence of primers 2113 and 2114, additional rounds of PCR can add the barcode region 2109 to the end of the target polynucleotide that attached to barcode region 2107. Additional downstream preparation steps (e.g., bulk amplification, size selection, etc.) are then performed as needed.
Example 16 Barcoding with Multiple Annealing and Looping-Based Amplification (MALBAC)
As shown in FIG. 22, a primer template 2200 comprising a barcode region is combined with agents 2201 necessary for PCR into a capsule 2202, using, for example interfacial polymerization or any other method described herein. PCR is then used to generate a primer from template 2200. Next, the capsule 2200 is encapsulated into an outer capsule 2003 that also comprises a mixture 2204 that comprises a target polynucleotide 2205 to be barcoded, reagents 2206 necessary for MALBAC amplification (e.g., DeepVent polymerase, dNTPs, buffer), and a MALBAC primer 2207 that does not comprise a barcode (similar to MALBAC primer 2100 described in Example 15). MALBAC amplification of the target polynucleotide 2205 commences to produce a MALBAC product similar to that described as that shown as 2110 in FIG. 21a . Capsule 2202 is then broken upon proper exposure of capsule 2202 to a stimulus designed to rupture capsule 2202, the contents of capsule 2202 mix with those of mixture 2004. A single cycle of PCR commences using the primer generated from template 2200 to generate a barcoded product, similar to that described in Example 15.
Outer capsule 2203 is then broken with the appropriate stimulus and the contents recovered. Additional downstream preparation steps (e.g., bulk amplification, size selection, addition of additional barcodes, etc.) are then performed as needed.
Example 17 Barcoding with Transposase and Tagmentation
As shown in FIG. 23, a single-stranded adapter-barcode polynucleotide sequence 2300 is synthesized, partitioned, amplified, and sorted as described in Example 1, or by any other method described in this disclosure. Interfacial polymerization is performed on the droplet comprising the single-stranded adapter-barcode polynucleotide sequence, to generate a capsule 2301.
Two mixtures are prepared. Mixture Z1 comprises a target polynucleotide 2302 (i.e., a polynucleotide to be fragmented and barcoded), a transposome 2303, and a partially complementary universal sequence 2304. A second mixture Z2 comprises capsule 2301, generated as described above and reagents 2305 necessary for PCR as described elsewhere herein.
Mixtures Z1 and Z2 are combined and a capsule within a capsule is formed according to methods described elsewhere in this disclosure, such as flow focusing. FIG. 23 illustrates a capsule within a capsule produced according to the method described above. The outer capsule 2306 comprises capsule 2301 and medium 2307. Capsule 2301 is one member of a library of encapsulated single-stranded adapter-barcode polynucleotides. Thus, capsule 2301 comprises multiple copies of a single-stranded adapter-barcode polynucleotide 2300, which can be used to attach the same barcode to a polynucleotide within a partition, such as outer capsule 2306.
The medium 2307 contains the contents of mixtures Z1 and Z2, described above. More specifically, medium 2307 comprises target polynucleotide 2302, the partially complementary universal sequence 2304, and the reagents 2305 necessary for PCR, including a hot start Taq.
Upon generation of the capsule within capsule, and exposure of the capsule within capsule to appropriate conditions, the transposome process the target polynucleotide. More specifically, the transposase fragments the target polynucleotide via tagmentations and tags it with a common priming sequence. The tagged target polynucleotide is then heated to fill in any gap in the target nucleotide generated by the transposase. The transposase is then heat inactivated and a stimulus is used to rupture inner capsule 2301, releasing its contents into outer capsule 2306. The hot start Taq is activated by heating the outer capsule 2306 to 95° C. The reaction proceeds with limited cycle PCR to add single-stranded adapter-barcode polynucleotide sequence 2300 to target polynucleotide 2302. The outer capsule 2306 is then ruptured and the target polynucleotides are sequenced.
It should be understood from the foregoing that, while particular implementations have been illustrated and described, various modifications may be made thereto and are contemplated herein. It is also not intended that the invention be limited by the specific examples provided within the specification. While the invention has been described with reference to the aforementioned specification, the descriptions and illustrations of the preferable embodiments herein are not meant to be construed in a limiting sense. Furthermore, it shall be understood that all aspects of the invention are not limited to the specific depictions, configurations or relative proportions set forth herein which depend upon a variety of conditions and variables. Various modifications in form and detail of the embodiments of the invention will be apparent to a person skilled in the art. It is therefore contemplated that the invention shall also cover any such modifications, variations and equivalents. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (40)

What is claimed is:
1. A method of synthesizing a library of polynucleotides comprising barcode sequences, said method comprising:
synthesizing a plurality of polynucleotides comprising barcode sequences;
separating said polynucleotides into a plurality of partitions, thereby generating partitioned polynucleotides, wherein each partition comprising a polynucleotide comprises multiple copies of the same polynucleotide comprising a barcode sequence prior to amplifying said partitioned polynucleotides;
amplifying said partitioned polynucleotides, thereby generating a library of amplified polynucleotides, comprising at least about 100,000 different barcode sequences;
wherein said amplified polynucleotides are attached to a bead; and
isolating partitions comprising amplified polynucleotides.
2. The method of claim 1, wherein said synthesizing comprises including a mixture of adenine, thymine, guanine, and cytosine in a coupling reaction.
3. The method of claim 1, wherein said separating comprises performing a limiting dilution, thereby generating diluted polynucleotides.
4. The method of claim 3, wherein said separating further comprises partitioning said diluted polynucleotides.
5. The method of claim 1, wherein said amplifying is performed by a method selected from the group consisting of polymerase chain reaction, asymmetric polymerase chain reaction, emulsion PCR (ePCR), ePCR including the use of a bead, ePCR including the use of a hydrogel, multiple annealing and looping-based amplification cycles (MALBAC), single primer isothermal amplification, and combinations thereof.
6. The method of claim 1, wherein said isolating is performed by flow-assisted sorting.
7. The method of claim 1, wherein a hairpin structure is formed from a polynucleotide selected from the group consisting of said polynucleotides comprising barcode sequences and said amplified polynucleotides.
8. The method of claim 7, further comprising cutting said hairpin structure within an unannealed region.
9. The method of claim 1, further comprising annealing said amplified polynucleotides with a partially complementary sequence.
10. The method of claim 9, wherein said partially complementary sequence comprises a barcode sequence.
11. The method of claim 1, further comprising attaching at least one of said amplified polynucleotides to a target sequence.
12. The method of claim 11, further comprising fragmenting said target sequence prior to said attaching step.
13. The method of claim 12, wherein fragmenting said target sequence is by a method selected from the group consisting of mechanical shear and treatment with an enzyme.
14. The method of claim 13, wherein said mechanical shear is induced by ultrasound.
15. The method of claim 13, wherein said enzyme is selected from the group consisting of a restriction enzyme, a fragmentase, and a transposase.
16. The method of claim 11, wherein said attaching is performed by a method selected from the group consisting of ligation and amplification.
17. The method of claim 16, wherein said amplification is a MALBAC amplification performed with MALBAC primers, thereby generating a MALBAC amplification product.
18. The method of claim 17, wherein said MALBAC primers comprise said amplified polynucleotides.
19. The method of claim 1, wherein following the separating step, the plurality of partitions comprises, on average, about 0.5 polynucleotides comprising barcode sequences per partition.
20. The method of claim 1, wherein following the separating step, the plurality of partitions comprises, on average, about 0.1 polynucleotides comprising barcode sequences per partition.
21. The method of claim 1, wherein said partitions are selected from the group consisting of droplets, capsules, and wells.
22. The method of claim 1, wherein said polynucleotides comprising barcode sequences further comprise a sequence selected from the group consisting of an immobilization sequence, an annealing sequence for a sequencing primer, and a sequence compatible for ligation with a target polynucleotide.
23. The method of claim 1, wherein said library comprises at least about 500,000 different barcode sequences.
24. The method of claim 1, wherein said library comprises at least about 1,000,000 different barcode sequences.
25. The method of claim 1, wherein said library comprises at least about 2,500,000 different barcode sequences.
26. The method of claim 1, wherein said library comprises at least about 5,000,000 different barcode sequences.
27. The method of claim 1, wherein said library comprises at least about 10,000,000 different barcode sequences.
28. The method of claim 1, wherein said library comprises at least about 25,000,000 different barcode sequences.
29. The method of claim 1, wherein said library comprises at least about 50,000,000 different barcode sequences.
30. The method of claim 1, wherein said library comprises at least about 100,000,000 different barcode sequences.
31. A method of synthesizing a library of polynucleotides comprising barcode sequences, said method comprising:
synthesizing a plurality of polynucleotides comprising barcode sequences;
separating said polynucleotides into a plurality of partitions, thereby generating partitioned polynucleotides;
amplifying said partitioned polynucleotides, using an RNA primer thereby generating amplified polynucleotides;
optionally exposing said amplified polynucleotides to RNase H; and
isolating partitions comprising amplified polynucleotides.
32. A method of synthesizing a library of polynucleotides comprising barcode sequences, said method comprising:
synthesizing a plurality of MALBAC primer polynucleotides comprising barcode sequences;
separating said polynucleotides into a plurality of partitions, thereby generating partitioned polynucleotides;
amplifying said partitioned polynucleotides, thereby generating amplified polynucleotides; and
isolating partitions comprising amplified polynucleotides.
33. The method of claim 31, wherein said library comprises at least about 500,000 different barcode sequences.
34. The method of claim 31, wherein said library comprises at least about 1,000,000 different barcode sequences.
35. The method of claim 31, wherein said library comprises at least about 2,500,000 different barcode sequences.
36. The method of claim 31, wherein said library comprises at least about 5,000,000 different barcode sequences.
37. The method of claim 32, wherein said library comprises at least about 500,000 different barcode sequences.
38. The method of claim 32, wherein said library comprises at least about 1,000,000 different barcode sequences.
39. The method of claim 32, wherein said library comprises at least about 2,500,000 different barcode sequences.
40. The method of claim 32, wherein said library comprises at least about 5,000,000 different barcode sequences.
US14/175,973 2012-08-14 2014-02-07 Polynucleotide barcode generation Active US9388465B2 (en)

Priority Applications (28)

Application Number Priority Date Filing Date Title
US14/175,973 US9388465B2 (en) 2013-02-08 2014-02-07 Polynucleotide barcode generation
US14/250,701 US20140235506A1 (en) 2013-02-08 2014-04-11 Polynucleotide barcode generation
US15/200,928 US20160304860A1 (en) 2013-02-08 2016-07-01 Polynucleotide barcode generation
US15/376,582 US9701998B2 (en) 2012-12-14 2016-12-12 Methods and systems for processing polynucleotides
US15/588,519 US9856530B2 (en) 2012-12-14 2017-05-05 Methods and systems for processing polynucleotides
US15/850,241 US10676789B2 (en) 2012-12-14 2017-12-21 Methods and systems for processing polynucleotides
US15/980,473 US10253364B2 (en) 2012-12-14 2018-05-15 Method and systems for processing polynucleotides
US16/000,803 US20180282804A1 (en) 2012-12-14 2018-06-05 Methods and systems for processing polynucleotides
US16/052,431 US10273541B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/052,486 US10323279B2 (en) 2012-08-14 2018-08-01 Methods and systems for processing polynucleotides
US16/165,389 US10533221B2 (en) 2012-12-14 2018-10-19 Methods and systems for processing polynucleotides
US16/212,441 US10752949B2 (en) 2012-08-14 2018-12-06 Methods and systems for processing polynucleotides
US16/231,185 US10400280B2 (en) 2012-08-14 2018-12-21 Methods and systems for processing polynucleotides
US16/231,142 US10584381B2 (en) 2012-08-14 2018-12-21 Methods and systems for processing polynucleotides
US16/246,322 US10597718B2 (en) 2012-08-14 2019-01-11 Methods and systems for sample processing polynucleotides
US16/294,769 US10450607B2 (en) 2012-08-14 2019-03-06 Methods and systems for processing polynucleotides
US16/395,090 US10669583B2 (en) 2012-08-14 2019-04-25 Method and systems for processing polynucleotides
US16/435,362 US10626458B2 (en) 2012-08-14 2019-06-07 Methods and systems for processing polynucleotides
US16/435,417 US10752950B2 (en) 2012-08-14 2019-06-07 Methods and systems for processing polynucleotides
US16/698,740 US11473138B2 (en) 2012-12-14 2019-11-27 Methods and systems for processing polynucleotides
US16/736,323 US20200255894A1 (en) 2012-08-14 2020-01-07 Methods and systems for sample processing polynucleotides
US16/844,141 US11441179B2 (en) 2012-08-14 2020-04-09 Methods and systems for processing polynucleotides
US16/852,906 US11421274B2 (en) 2012-12-14 2020-04-20 Methods and systems for processing polynucleotides
US16/998,414 US11021749B2 (en) 2012-08-14 2020-08-20 Methods and systems for processing polynucleotides
US16/998,425 US11035002B2 (en) 2012-08-14 2020-08-20 Methods and systems for processing polynucleotides
US17/314,526 US11359239B2 (en) 2012-08-14 2021-05-07 Methods and systems for processing polynucleotides
US17/392,610 US20220098659A1 (en) 2012-08-14 2021-08-03 Methods and systems for processing polynucleotides
US18/186,088 US20240002929A1 (en) 2012-08-14 2023-03-17 Methods and systems for processing polynucleotides

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201361762435P 2013-02-08 2013-02-08
US201361800223P 2013-03-15 2013-03-15
US201361840403P 2013-06-27 2013-06-27
US201361844804P 2013-07-10 2013-07-10
US14/175,973 US9388465B2 (en) 2013-02-08 2014-02-07 Polynucleotide barcode generation

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/598,898 Continuation-In-Part US20170321252A1 (en) 2012-08-14 2017-05-18 Capsule array devices and methods of use
US15/847,752 Continuation-In-Part US10480028B2 (en) 2012-08-14 2017-12-19 Methods and systems for processing polynucleotides

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/250,701 Continuation US20140235506A1 (en) 2012-08-14 2014-04-11 Polynucleotide barcode generation

Publications (2)

Publication Number Publication Date
US20140228255A1 US20140228255A1 (en) 2014-08-14
US9388465B2 true US9388465B2 (en) 2016-07-12

Family

ID=51297676

Family Applications (8)

Application Number Title Priority Date Filing Date
US14/175,935 Active 2034-04-09 US9644204B2 (en) 2013-02-08 2014-02-07 Partitioning and processing of analytes and other species
US14/175,973 Active US9388465B2 (en) 2012-08-14 2014-02-07 Polynucleotide barcode generation
US14/250,701 Abandoned US20140235506A1 (en) 2012-08-14 2014-04-11 Polynucleotide barcode generation
US15/200,928 Abandoned US20160304860A1 (en) 2012-12-14 2016-07-01 Polynucleotide barcode generation
US15/470,814 Active US10150963B2 (en) 2013-02-08 2017-03-27 Partitioning and processing of analytes and other species
US15/693,374 Active US10150964B2 (en) 2013-02-08 2017-08-31 Partitioning and processing of analytes and other species
US16/170,980 Active 2034-06-07 US11193121B2 (en) 2013-02-08 2018-10-25 Partitioning and processing of analytes and other species
US17/517,287 Pending US20220154175A1 (en) 2013-02-08 2021-11-02 Partitioning and processing of analytes and other species

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/175,935 Active 2034-04-09 US9644204B2 (en) 2013-02-08 2014-02-07 Partitioning and processing of analytes and other species

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/250,701 Abandoned US20140235506A1 (en) 2012-08-14 2014-04-11 Polynucleotide barcode generation
US15/200,928 Abandoned US20160304860A1 (en) 2012-12-14 2016-07-01 Polynucleotide barcode generation
US15/470,814 Active US10150963B2 (en) 2013-02-08 2017-03-27 Partitioning and processing of analytes and other species
US15/693,374 Active US10150964B2 (en) 2013-02-08 2017-08-31 Partitioning and processing of analytes and other species
US16/170,980 Active 2034-06-07 US11193121B2 (en) 2013-02-08 2018-10-25 Partitioning and processing of analytes and other species
US17/517,287 Pending US20220154175A1 (en) 2013-02-08 2021-11-02 Partitioning and processing of analytes and other species

Country Status (10)

Country Link
US (8) US9644204B2 (en)
EP (4) EP2954065B1 (en)
JP (4) JP2016511243A (en)
KR (3) KR20230003659A (en)
CN (2) CN108753766A (en)
AU (1) AU2014214682B2 (en)
BR (1) BR112015019159A2 (en)
CA (2) CA2900481A1 (en)
DK (1) DK2954065T3 (en)
WO (2) WO2014124338A1 (en)

Cited By (84)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9644204B2 (en) 2013-02-08 2017-05-09 10X Genomics, Inc. Partitioning and processing of analytes and other species
US9689024B2 (en) 2012-08-14 2017-06-27 10X Genomics, Inc. Methods for droplet-based sample preparation
US9694361B2 (en) 2014-04-10 2017-07-04 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10106789B2 (en) 2010-12-16 2018-10-23 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
WO2019032760A1 (en) 2017-08-10 2019-02-14 Rootpath Genomics, Inc. Improved method to analyze nucleic acid contents from multiple biological particles
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10227648B2 (en) 2012-12-14 2019-03-12 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10287623B2 (en) 2014-10-29 2019-05-14 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10357771B2 (en) 2017-08-22 2019-07-23 10X Genomics, Inc. Method of producing emulsions
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10544413B2 (en) 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10590244B2 (en) 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10596541B2 (en) 2014-04-21 2020-03-24 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
US10650912B2 (en) 2015-01-13 2020-05-12 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US10725027B2 (en) 2018-02-12 2020-07-28 10X Genomics, Inc. Methods and systems for analysis of chromatin
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
WO2020207963A1 (en) 2019-04-12 2020-10-15 Miltenyi Biotec B.V. & Co. KG Conjugates having an enzymatically releasable detection moiety and a barcode moiety
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US10839939B2 (en) 2014-06-26 2020-11-17 10X Genomics, Inc. Processes and systems for nucleic acid sequence assembly
US10837047B2 (en) 2017-10-04 2020-11-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10854315B2 (en) 2015-02-09 2020-12-01 10X Genomics, Inc. Systems and methods for determining structural variation and phasing using variant call data
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
US11001883B2 (en) 2012-03-05 2021-05-11 The General Hospital Corporation Systems and methods for epigenetic sequencing
WO2021094421A1 (en) 2019-11-15 2021-05-20 Miltenyi Biotec B.V. & Co. KG Color and bardcoded beads for single cell indexing
US11072816B2 (en) 2017-05-03 2021-07-27 The Broad Institute, Inc. Single-cell proteomic assay using aptamers
US11081208B2 (en) 2016-02-11 2021-08-03 10X Genomics, Inc. Systems, methods, and media for de novo assembly of whole genome sequence data
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US20210254051A1 (en) * 2018-04-03 2021-08-19 Guangzhou Burning Rock Dx Co., Ltd. Compositions and methods for preparing nucleic acid libraries
US11123735B2 (en) 2019-10-10 2021-09-21 1859, Inc. Methods and systems for microfluidic screening
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
WO2022048780A1 (en) 2020-09-07 2022-03-10 Miltenyi Biotec B.V. & Co. KG Conjugates having an enzymatically releasable detection moiety and a barcode moiety
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
US11358137B2 (en) 2018-12-26 2022-06-14 Industrial Technology Research Institute Tubular structure for producing droplets and method for producing droplets
US11365438B2 (en) 2017-11-30 2022-06-21 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
US11371094B2 (en) 2015-11-19 2022-06-28 10X Genomics, Inc. Systems and methods for nucleic acid processing using degenerate nucleotides
US11459607B1 (en) 2018-12-10 2022-10-04 10X Genomics, Inc. Systems and methods for processing-nucleic acid molecules from a single cell using sequential co-partitioning and composite barcodes
US11467153B2 (en) 2019-02-12 2022-10-11 10X Genomics, Inc. Methods for processing nucleic acid molecules
US11512337B2 (en) 2020-01-13 2022-11-29 Fluent Biosciences Inc. Emulsion based drug screening
US20220411859A1 (en) * 2017-08-01 2022-12-29 Illumina, Inc. Hydrogel beads for nucleotide sequencing
US11566279B2 (en) 2014-09-09 2023-01-31 The Broad Institute, Inc. Droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11584953B2 (en) 2019-02-12 2023-02-21 10X Genomics, Inc. Methods for processing nucleic acid molecules
US11584954B2 (en) 2017-10-27 2023-02-21 10X Genomics, Inc. Methods and systems for sample preparation and analysis
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
US11655499B1 (en) 2019-02-25 2023-05-23 10X Genomics, Inc. Detection of sequence elements in nucleic acid molecules
US11660601B2 (en) 2017-05-18 2023-05-30 10X Genomics, Inc. Methods for sorting particles
US11667951B2 (en) 2016-10-24 2023-06-06 Geneinfosec, Inc. Concealing information present within nucleic acids
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
US11725231B2 (en) 2017-10-26 2023-08-15 10X Genomics, Inc. Methods and systems for nucleic acid preparation and chromatin analysis
US11746367B2 (en) 2015-04-17 2023-09-05 President And Fellows Of Harvard College Barcoding systems and methods for gene sequencing and other applications
US11773452B2 (en) 2020-01-13 2023-10-03 Fluent Biosciences Inc. Single cell sequencing
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11827936B2 (en) 2020-01-13 2023-11-28 Fluent Biosciences Inc. Methods and systems for single cell gene profiling
US11833515B2 (en) 2017-10-26 2023-12-05 10X Genomics, Inc. Microfluidic channel networks for partitioning
US11845983B1 (en) 2019-01-09 2023-12-19 10X Genomics, Inc. Methods and systems for multiplexing of droplet based assays
US11851700B1 (en) 2020-05-13 2023-12-26 10X Genomics, Inc. Methods, kits, and compositions for processing extracellular molecules
US11851683B1 (en) 2019-02-12 2023-12-26 10X Genomics, Inc. Methods and systems for selective analysis of cellular samples
US11866782B2 (en) 2020-03-16 2024-01-09 Fluent Biosciences Inc. Multi-omic analysis in monodisperse droplets
US11873530B1 (en) 2018-07-27 2024-01-16 10X Genomics, Inc. Systems and methods for metabolome analysis
US11873483B2 (en) 2015-03-11 2024-01-16 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
US11898206B2 (en) 2017-05-19 2024-02-13 10X Genomics, Inc. Systems and methods for clonotype screening
US11904310B2 (en) 2015-10-28 2024-02-20 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
US11920183B2 (en) 2019-03-11 2024-03-05 10X Genomics, Inc. Systems and methods for processing optically tagged beads
US11932899B2 (en) 2018-06-07 2024-03-19 10X Genomics, Inc. Methods and systems for characterizing nucleic acid molecules
US11952626B2 (en) 2023-08-22 2024-04-09 10X Genomics, Inc. Probe-based analysis of nucleic acids and proteins

Families Citing this family (146)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008109176A2 (en) 2007-03-07 2008-09-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
WO2009085215A1 (en) 2007-12-21 2009-07-09 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
EP2373812B1 (en) 2008-12-19 2016-11-09 President and Fellows of Harvard College Particle-assisted nucleic acid sequencing
EP2406003A2 (en) 2009-03-13 2012-01-18 President and Fellows of Harvard College Scale-up of flow-focusing microfluidic devices
KR20120089661A (en) 2009-09-02 2012-08-13 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Multiple emulsions created using jetting and other techniques
CN102648053B (en) 2009-10-27 2016-04-27 哈佛学院院长等 Drop formation technology
US9315857B2 (en) 2009-12-15 2016-04-19 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse label-tags
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
JP2014512826A (en) 2011-04-25 2014-05-29 バイオ−ラド ラボラトリーズ インコーポレイテッド Methods and compositions for nucleic acid analysis
EP2714254B1 (en) 2011-05-23 2017-09-06 President and Fellows of Harvard College Control of emulsions, including multiple emulsions
EP2820174B1 (en) 2012-02-27 2019-12-25 The University of North Carolina at Chapel Hill Methods and uses for molecular tags
EP3321378B1 (en) 2012-02-27 2021-11-10 Becton, Dickinson and Company Compositions for molecular counting
WO2014142850A1 (en) 2013-03-13 2014-09-18 Illumina, Inc. Methods and compositions for nucleic acid sequencing
EP3013983B1 (en) 2013-06-25 2023-02-15 Prognosys Biosciences, Inc. Spatially encoded biological assays using a microfluidic device
KR102536833B1 (en) 2013-08-28 2023-05-26 벡톤 디킨슨 앤드 컴퍼니 Massively parallel single cell analysis
CN105745528A (en) 2013-10-07 2016-07-06 赛卢拉研究公司 Methods and systems for digitally counting features on arrays
WO2015069634A1 (en) 2013-11-08 2015-05-14 President And Fellows Of Harvard College Microparticles, methods for their preparation and use
US11155809B2 (en) 2014-06-24 2021-10-26 Bio-Rad Laboratories, Inc. Digital PCR barcoding
US10017759B2 (en) * 2014-06-26 2018-07-10 Illumina, Inc. Library preparation of tagged nucleic acid
US11085084B2 (en) 2014-09-12 2021-08-10 The Board Of Trustees Of The Leland Stanford Junior University Identification and use of circulating nucleic acids
BR122021026779B1 (en) * 2014-10-17 2023-12-19 Illumina Cambridge Limited CONTIGUITY PRESERVING TRANSPOSON
US11119098B2 (en) 2014-10-31 2021-09-14 Massachusetts Institute Of Technology Systems including Janus droplets
US10005058B2 (en) * 2014-10-31 2018-06-26 Massachusetts Institute Of Technology Compositions and methods for arranging colloid phases
US10060913B2 (en) 2016-09-19 2018-08-28 Massachusetts Institute Of Technology Systems including janus droplets capable of binding an analyte and changing orientation to provide a detectable change
EP3212315A4 (en) 2014-10-31 2018-07-11 Massachusetts Institute of Technology Compositions and methods for forming emulsions
US10900065B2 (en) 2014-11-14 2021-01-26 University Of Washington Methods and kits for labeling cellular molecules
KR20210135626A (en) * 2015-02-10 2021-11-15 일루미나, 인코포레이티드 The method and the composition for analyzing the cellular constituent
CA2974398A1 (en) * 2015-02-13 2016-08-18 Abhijit Ajit PATEL Methods for highly parallel and accurate measurement of nucleic acids
WO2016134078A1 (en) 2015-02-19 2016-08-25 Becton, Dickinson And Company High-throughput single-cell analysis combining proteomic and genomic information
CN107208158B (en) 2015-02-27 2022-01-28 贝克顿迪金森公司 Spatially addressable molecular barcode
WO2016145416A2 (en) * 2015-03-11 2016-09-15 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
EP3268125A4 (en) 2015-03-13 2018-08-15 President and Fellows of Harvard College Determination of cells using amplification
CN107430646B (en) 2015-03-17 2021-10-22 生物辐射实验室股份有限公司 Detecting genome editing
WO2016149418A1 (en) * 2015-03-18 2016-09-22 Cellular Research, Inc. Methods and compositions for labeling targets and haplotype phasing
EP4180535A1 (en) 2015-03-30 2023-05-17 Becton, Dickinson and Company Methods and compositions for combinatorial barcoding
FI3901281T3 (en) 2015-04-10 2023-01-31 Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11661597B2 (en) * 2015-04-15 2023-05-30 The Board Of Trustees Of The Leland Stanford Junior University Robust quantification of single molecules in next-generation sequencing using non-random combinatorial oligonucleotide barcodes
EP3285926B1 (en) * 2015-04-21 2022-03-02 General Automation Lab Technologies Inc. Kit and method for high throughput microbiology applications
WO2016172373A1 (en) 2015-04-23 2016-10-27 Cellular Research, Inc. Methods and compositions for whole transcriptome amplification
DK3294911T3 (en) 2015-05-11 2020-11-16 Illumina Inc Platform for discovery and analysis of therapeutic agents
US11124823B2 (en) 2015-06-01 2021-09-21 Becton, Dickinson And Company Methods for RNA quantification
US9771575B2 (en) 2015-06-19 2017-09-26 Agilent Technologies, Inc. Methods for on-array fragmentation and barcoding of DNA samples
US10640816B2 (en) 2015-07-17 2020-05-05 Nanostring Technologies, Inc. Simultaneous quantification of gene expression in a user-defined region of a cross-sectioned tissue
CN108026524A (en) 2015-09-11 2018-05-11 赛卢拉研究公司 Method and composition for nucleic acid library standardization
US11123297B2 (en) 2015-10-13 2021-09-21 President And Fellows Of Harvard College Systems and methods for making and using gel microspheres
WO2017070056A1 (en) 2015-10-20 2017-04-27 10X Genomics, Inc. Methods and systems for high throughput single cell genetic manipulation
EP3368668B1 (en) * 2015-10-28 2023-11-29 Silicon Valley Scientific, Inc. Method and apparatus for encoding cellular spatial position information
SG11201803983UA (en) * 2015-11-19 2018-06-28 10X Genomics Inc Transformable tagging compositions, methods, and processes incorporating same
WO2017100350A1 (en) * 2015-12-07 2017-06-15 Raindance Technologies, Inc. Multiplexing in partitions using microparticles
CN105567562B (en) * 2015-12-30 2017-10-03 中国科学院合肥物质科学研究院 Microfluid system and its introduction method for unicellular ultrasonic wave channel genes
US10633648B2 (en) 2016-02-12 2020-04-28 University Of Washington Combinatorial photo-controlled spatial sequencing and labeling
US20170283864A1 (en) * 2016-03-31 2017-10-05 Agilent Technologies, Inc. Use of transposase and y adapters to fragment and tag dna
WO2017173328A1 (en) * 2016-04-01 2017-10-05 Baylor College Of Medicine Methods of whole transcriptome amplification
CA3021735A1 (en) * 2016-04-19 2017-10-26 President And Fellows Of Harvard College Immobilization-based systems and methods for genetic analysis and other applications
AU2017261189B2 (en) 2016-05-02 2023-02-09 Becton, Dickinson And Company Accurate molecular barcoding
US10301677B2 (en) 2016-05-25 2019-05-28 Cellular Research, Inc. Normalization of nucleic acid libraries
JPWO2017204294A1 (en) * 2016-05-25 2019-02-21 株式会社ニコン Target biomolecule identification method, target biomolecule identification beads, bead set, and target biomolecule identification apparatus
US11397882B2 (en) 2016-05-26 2022-07-26 Becton, Dickinson And Company Molecular label counting adjustment methods
US10640763B2 (en) 2016-05-31 2020-05-05 Cellular Research, Inc. Molecular indexing of internal sequences
US10202641B2 (en) 2016-05-31 2019-02-12 Cellular Research, Inc. Error correction in amplification of samples
US10676736B2 (en) 2016-08-05 2020-06-09 Bio-Rad Laboratories, Inc. Second strand direct
RU2019106038A (en) * 2016-08-10 2020-09-17 Президент Энд Фэллоуз Оф Харвард Коллидж DE NOVO METHODS FOR ASSEMBLING BARCODED GENOMIC DNA FRAGMENTS
US10688494B2 (en) 2016-08-23 2020-06-23 10X Genomics, Inc. Microfluidic surface-mediated emulsion stability control
US11460468B2 (en) 2016-09-26 2022-10-04 Becton, Dickinson And Company Measurement of protein expression using reagents with barcoded oligonucleotide sequences
KR20190077062A (en) 2016-11-08 2019-07-02 셀룰러 리서치, 인크. How to classify expression profiles
EP3538672A1 (en) 2016-11-08 2019-09-18 Cellular Research, Inc. Methods for cell label classification
US20200190507A1 (en) * 2016-11-10 2020-06-18 The Scripps Research Institute Encoded Solid Phase Compound Library with Polynucleotide Based Barcoding
WO2018118971A1 (en) 2016-12-19 2018-06-28 Bio-Rad Laboratories, Inc. Droplet tagging contiguity preserved tagmented dna
CN110573253B (en) 2017-01-13 2021-11-02 赛卢拉研究公司 Hydrophilic coating for fluid channels
EP3574112B1 (en) 2017-01-27 2023-01-11 Roche Diagnostics GmbH Barcoded dna for long range sequencing
US11319583B2 (en) 2017-02-01 2022-05-03 Becton, Dickinson And Company Selective amplification using blocking oligonucleotides
EP3391958B1 (en) * 2017-04-19 2020-08-12 The Procter & Gamble Company Method of making surface-coated water-absorbing polymer particles in a microfluidic device
EP3619325B1 (en) 2017-05-05 2024-01-24 Scipio Bioscience Methods for trapping and barcoding discrete biological units in hydrogel
WO2018226546A1 (en) 2017-06-05 2018-12-13 10X Genomics, Inc. Gaskets for the distribution of pressures in a microfluidic system
US10676779B2 (en) 2017-06-05 2020-06-09 Becton, Dickinson And Company Sample indexing for single cells
SG11201912824YA (en) 2017-06-20 2020-01-30 10X Genomics Inc Methods and systems for improved droplet stabilization
CN111356772B (en) * 2017-08-23 2023-10-03 豪夫迈·罗氏有限公司 Enzyme screening method
US10636761B2 (en) * 2017-08-29 2020-04-28 Electronics And Telecommunications Reearch Institute Method of fabricating a semiconductor package
US10748643B2 (en) 2017-08-31 2020-08-18 10X Genomics, Inc. Systems and methods for determining the integrity of test strings with respect to a ground truth string
CN109486923B (en) * 2017-09-11 2022-02-18 广州永诺生物科技有限公司 Primer system for sequencing multiple amplicons, application thereof and method for constructing sequencing library
WO2019074524A1 (en) 2017-10-13 2019-04-18 Hewlett-Packard Development Company, L.P. Partition liquid into samples
EP4180534A1 (en) 2017-11-02 2023-05-17 Bio-Rad Laboratories, Inc. Transposase-based genomic analysis
JP7049103B2 (en) * 2017-12-11 2022-04-06 株式会社日立製作所 Comprehensive 3'end gene expression analysis method for single cells
US11946095B2 (en) 2017-12-19 2024-04-02 Becton, Dickinson And Company Particles associated with oligonucleotides
WO2019148042A1 (en) * 2018-01-26 2019-08-01 10X Genomics, Inc. Compositions and methods for sample processing
CN111699253A (en) 2018-01-31 2020-09-22 生物辐射实验室股份有限公司 Methods and compositions for deconvolving a partitioned barcode
US20190249248A1 (en) 2018-02-12 2019-08-15 Nanostring Technologies, Inc. Biomolecular probes and methods of detecting gene and protein expression
CA3097976A1 (en) 2018-05-03 2019-11-07 Becton, Dickinson And Company High throughput multiomics sample analysis
WO2019213237A1 (en) 2018-05-03 2019-11-07 Becton, Dickinson And Company Molecular barcoding on opposite transcript ends
DE102018207098A1 (en) * 2018-05-08 2019-11-14 Robert Bosch Gmbh Microfluidic device and method for nanostructure sequencing of nucleotide strands
WO2019222567A1 (en) 2018-05-18 2019-11-21 Massachusetts Institute Of Technology Light emitting droplets and related methods
US11479816B2 (en) 2018-08-20 2022-10-25 Bio-Rad Laboratories, Inc. Nucleotide sequence generation by barcode bead-colocalization in partitions
JP2022511398A (en) 2018-10-01 2022-01-31 ベクトン・ディキンソン・アンド・カンパニー Determining the 5'transcription sequence
US11932849B2 (en) 2018-11-08 2024-03-19 Becton, Dickinson And Company Whole transcriptome analysis of single cells using random priming
CN113195717A (en) 2018-12-13 2021-07-30 贝克顿迪金森公司 Selective extension in single cell whole transcriptome analysis
US11926867B2 (en) 2019-01-06 2024-03-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
WO2020150356A1 (en) 2019-01-16 2020-07-23 Becton, Dickinson And Company Polymerase chain reaction normalization through primer titration
WO2020154247A1 (en) 2019-01-23 2020-07-30 Cellular Research, Inc. Oligonucleotides associated with antibodies
EP3923977A1 (en) 2019-02-12 2021-12-22 TheryCell GmbH Reverse immunosuppression
US20220072535A1 (en) * 2019-04-30 2022-03-10 Hewlett-Packard Development Company, L.P. Microfluidic devices
CN114051534A (en) 2019-07-22 2022-02-15 贝克顿迪金森公司 Single cell chromatin immunoprecipitation sequencing assay
JP2022544662A (en) * 2019-08-19 2022-10-20 ユニバーサル シーケンシング テクノロジー コーポレイション Methods and Compositions for Tracking Nucleic Acid Fragment Origins for Nucleic Acid Sequencing
WO2021092433A2 (en) 2019-11-08 2021-05-14 10X Genomics, Inc. Enhancing specificity of analyte binding
EP4055185A1 (en) 2019-11-08 2022-09-14 10X Genomics, Inc. Spatially-tagged analyte capture agents for analyte multiplexing
EP4055160B1 (en) 2019-11-08 2024-04-10 Becton Dickinson and Company Using random priming to obtain full-length v(d)j information for immune repertoire sequencing
US11857981B2 (en) 2019-12-23 2024-01-02 10X Genomics, Inc. Magnetic separator for an automated single cell sequencing system
CN114885610A (en) 2019-12-23 2022-08-09 10X基因组学有限公司 Methods for spatial analysis using RNA templated ligation
US11649497B2 (en) 2020-01-13 2023-05-16 Becton, Dickinson And Company Methods and compositions for quantitation of proteins and RNA
US11702693B2 (en) 2020-01-21 2023-07-18 10X Genomics, Inc. Methods for printing cells and generating arrays of barcoded cells
US11732299B2 (en) 2020-01-21 2023-08-22 10X Genomics, Inc. Spatial assays with perturbed cells
US11821035B1 (en) 2020-01-29 2023-11-21 10X Genomics, Inc. Compositions and methods of making gene expression libraries
US11898205B2 (en) 2020-02-03 2024-02-13 10X Genomics, Inc. Increasing capture efficiency of spatial assays
EP3859012A1 (en) * 2020-02-03 2021-08-04 Albert-Ludwigs-Universität Freiburg Methods for amplification of genomic dna and preparation of sequencing libraries
US11732300B2 (en) 2020-02-05 2023-08-22 10X Genomics, Inc. Increasing efficiency of spatial analysis in a biological sample
US11835462B2 (en) 2020-02-11 2023-12-05 10X Genomics, Inc. Methods and compositions for partitioning a biological sample
US11891654B2 (en) 2020-02-24 2024-02-06 10X Genomics, Inc. Methods of making gene expression libraries
US11926863B1 (en) 2020-02-27 2024-03-12 10X Genomics, Inc. Solid state single cell method for analyzing fixed biological cells
EP4110941A1 (en) 2020-02-28 2023-01-04 10X Genomics, Inc. Method for isolating nuclei and cells from tissues
US11768175B1 (en) 2020-03-04 2023-09-26 10X Genomics, Inc. Electrophoretic methods for spatial analysis
EP4136227A1 (en) 2020-04-16 2023-02-22 10X Genomics, Inc. Compositions and methods for use with fixed samples
EP4242325A3 (en) 2020-04-22 2023-10-04 10X Genomics, Inc. Methods for spatial analysis using targeted rna depletion
US11701668B1 (en) 2020-05-08 2023-07-18 10X Genomics, Inc. Methods and devices for magnetic separation
CN115605614A (en) 2020-05-14 2023-01-13 贝克顿迪金森公司(Us) Primers for immune repertoire profiling
WO2021237087A1 (en) 2020-05-22 2021-11-25 10X Genomics, Inc. Spatial analysis to detect sequence variants
EP4153775A1 (en) 2020-05-22 2023-03-29 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
US11946038B1 (en) 2020-05-29 2024-04-02 10X Genomics, Inc. Methods and systems including flow and magnetic modules
CN116249785A (en) 2020-06-02 2023-06-09 10X基因组学有限公司 Space transcriptomics for antigen-receptor
AU2021283174A1 (en) 2020-06-02 2023-01-05 10X Genomics, Inc. Nucleic acid library methods
EP4162074A1 (en) 2020-06-08 2023-04-12 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
EP4172362A1 (en) 2020-06-25 2023-05-03 10X Genomics, Inc. Spatial analysis of dna methylation
EP4176437A1 (en) 2020-07-02 2023-05-10 10X Genomics, Inc. Systems and methods for detection of low-abundance molecular barcodes from a sequencing library
US11761038B1 (en) 2020-07-06 2023-09-19 10X Genomics, Inc. Methods for identifying a location of an RNA in a biological sample
US11932901B2 (en) 2020-07-13 2024-03-19 Becton, Dickinson And Company Target enrichment using nucleic acid probes for scRNAseq
JP2023534028A (en) * 2020-07-15 2023-08-07 フルーエント バイオサイエンシーズ インコーポレイテッド Oligos for stepwise ligation
US11926822B1 (en) 2020-09-23 2024-03-12 10X Genomics, Inc. Three-dimensional spatial analysis
US11827935B1 (en) 2020-11-19 2023-11-28 10X Genomics, Inc. Methods for spatial analysis using rolling circle amplification and detection probes
EP4247967A1 (en) 2020-11-20 2023-09-27 Becton, Dickinson and Company Profiling of highly expressed and lowly expressed proteins
WO2022140028A1 (en) 2020-12-21 2022-06-30 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
AU2022238446A1 (en) 2021-03-18 2023-09-07 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample
WO2023014303A2 (en) * 2021-08-05 2023-02-09 Agency For Science, Technology And Research A method of amplification of a nucleic acid
WO2023034489A1 (en) 2021-09-01 2023-03-09 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
EP4272764A1 (en) 2022-05-03 2023-11-08 Scipio Bioscience Method of complexing biological units with particles
KR102578493B1 (en) 2023-04-26 2023-09-14 대한민국 A loading rack

Citations (208)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0249007A2 (en) 1986-04-14 1987-12-16 The General Hospital Corporation A method of screening hybridomas
US5149625A (en) 1987-08-11 1992-09-22 President And Fellows Of Harvard College Multiplex analysis of DNA
US5202231A (en) 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US5413924A (en) 1992-02-13 1995-05-09 Kosak; Kenneth M. Preparation of wax beads containing a reagent for release by heating
US5436130A (en) 1992-03-19 1995-07-25 The Regents Of The University Of California Multiple tag labeling method for DNA sequencing
US5512131A (en) 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
WO1996029629A2 (en) 1995-03-01 1996-09-26 President And Fellows Of Harvard College Microcontact printing on surfaces and derivative articles
WO1996041011A1 (en) 1995-06-07 1996-12-19 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
US5618711A (en) 1986-08-22 1997-04-08 Hoffmann-La Roche Inc. Recombinant expression vectors and purification methods for Thermus thermophilus DNA polymerase
EP0637996B1 (en) 1992-05-01 1997-07-23 The Trustees Of The University Of Pennsylvania Microfabricated detection structures
US5695940A (en) 1987-04-01 1997-12-09 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US5736330A (en) 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US5834197A (en) 1994-05-11 1998-11-10 Genera Technologies Limited Methods of capturing species from liquids and assay procedures
US5851769A (en) 1995-09-27 1998-12-22 The Regents Of The University Of California Quantitative DNA fiber mapping
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
WO1999009217A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US5958703A (en) 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
WO1999052708A1 (en) 1998-04-13 1999-10-21 Luminex Corporation Liquid labeling with fluorescent microparticles
US5994056A (en) 1991-05-02 1999-11-30 Roche Molecular Systems, Inc. Homogeneous methods for nucleic acid amplification and detection
WO2000008212A1 (en) 1998-08-07 2000-02-17 Cellay, Llc Gel microdrops in genetic analysis
US6046003A (en) 1995-11-30 2000-04-04 Pharmaseq, Inc. Method of determining the sequence of nucleic acids employing solid-phase particles carrying transponders
US6051377A (en) 1995-11-30 2000-04-18 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
WO2000026412A1 (en) 1998-11-02 2000-05-11 Kenneth Loren Beattie Nucleic acid analysis using sequence-targeted tandem hybridization
US6103537A (en) 1997-10-02 2000-08-15 Aclara Biosciences, Inc. Capillary assays involving separation of free and bound species
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
US6172218B1 (en) 1994-10-13 2001-01-09 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
WO2001014589A2 (en) 1999-08-20 2001-03-01 Luminex Corporation Liquid array technology
US20010020588A1 (en) 1997-09-15 2001-09-13 Whitehead Institute For Biomedical Research Methods and apparatus for processing a sample of biomolecular analyte using a microfabricated device
US6297006B1 (en) 1997-01-16 2001-10-02 Hyseq, Inc. Methods for sequencing repetitive sequences and for determining the order of sequence subfragments
US6297017B1 (en) 1997-07-11 2001-10-02 Brax Group Limited Categorising nucleic acids
WO2001089787A2 (en) 2000-05-25 2001-11-29 President And Fellows Of Harvard College Microfluidic systems including three-dimensionally arrayed channel networks
US6327410B1 (en) 1997-03-14 2001-12-04 The Trustees Of Tufts College Target analyte sensors utilizing Microspheres
US6355198B1 (en) 1996-03-15 2002-03-12 President And Fellows Of Harvard College Method of forming articles including waveguides via capillary micromolding and microtransfer molding
US20020034737A1 (en) 1997-03-04 2002-03-21 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US6361950B1 (en) 1995-11-30 2002-03-26 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
US6372813B1 (en) 1999-06-25 2002-04-16 Motorola Methods and compositions for attachment of biomolecules to solid supports, hydrogels, and hydrogel arrays
WO2002031203A2 (en) 2000-10-10 2002-04-18 Diversa Corporation High throughput or capillary-based screening for a bioactivity or biomolecule
US20020051992A1 (en) 1997-05-23 2002-05-02 Lynx Therapeutics, Inc. System and apparatus for sequential processing of analytes
US20020089100A1 (en) 2000-11-14 2002-07-11 Akira Kawasaki Production apparatus of monodisperse particle and production process of monodisperse particle and monodisperse particle produced by the process
US20020092767A1 (en) 1997-09-19 2002-07-18 Aclara Biosciences, Inc. Multiple array microfluidic device units
US6432360B1 (en) 1997-10-10 2002-08-13 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
WO2002086148A1 (en) 2001-04-18 2002-10-31 Ambrigen, Llc Particle based assay system
US6485944B1 (en) 1997-10-10 2002-11-26 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US20020179849A1 (en) 1999-05-12 2002-12-05 Kevin Maher Multiplexed fluorescent detection in microfluidic devices
US20030008285A1 (en) 2001-06-29 2003-01-09 Fischer Steven M. Method of DNA sequencing using cleavable tags
US20030008323A1 (en) 1999-04-15 2003-01-09 Ilya Ravkin Chemical-library composition and method
US6511803B1 (en) 1997-10-10 2003-01-28 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US20030028981A1 (en) 1997-10-14 2003-02-13 Chandler Don J. Precision fluorescently dyed particles and methods of making and using same
US6524456B1 (en) 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
US20030039978A1 (en) 2001-08-27 2003-02-27 Hannah Eric C. Electron induced fluorescent method for nucleic acid sequencing
US20030044836A1 (en) 1998-10-15 2003-03-06 Princeton University, Office Of Technology & Trademark Licensing Quantitative analysis of hybridization patterns and intensities in oligonucleotide arrays
US20030044777A1 (en) 1993-10-28 2003-03-06 Kenneth L. Beattie Flowthrough devices for multiple discrete binding reactions
US20030104466A1 (en) 1997-04-04 2003-06-05 Caliper Technologies Corporation Microfluidic sequencing systems
US20030170698A1 (en) 2002-01-04 2003-09-11 Peter Gascoyne Droplet-based microfluidic oligonucleotide synthesis engine
US20030182068A1 (en) 2001-10-30 2003-09-25 Battersby Bronwyn J. Device and methods for directed synthesis of chemical libraries
US6632655B1 (en) 1999-02-23 2003-10-14 Caliper Technologies Corp. Manipulation of microparticles in microfluidic systems
US6632606B1 (en) 2000-06-12 2003-10-14 Aclara Biosciences, Inc. Methods for single nucleotide polymorphism detection
WO2004002627A2 (en) 2002-06-28 2004-01-08 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
WO2004010106A2 (en) 2002-07-24 2004-01-29 Ptc Therapeutics, Inc. METHODS FOR IDENTIFYING SMALL MOLEDULES THAT MODULATE PREMATURE TRANSLATION TERMINATION AND NONSENSE MEDIATED mRNA DECAY
US20040063138A1 (en) 1999-02-16 2004-04-01 Mcginnis Malcolm D. Polynucleotide sequencing method
US20040132122A1 (en) 2000-06-21 2004-07-08 Sukanta Banerjee Multianalyte molecular analysis using application-specific random particle arrays
EP1019496B1 (en) 1997-07-07 2004-09-29 Medical Research Council In vitro sorting method
US6800298B1 (en) 2000-05-11 2004-10-05 Clemson University Biological lubricant composition and method of applying lubricant composition
US6806058B2 (en) 2001-05-26 2004-10-19 One Cell Systems, Inc. Secretions of proteins by encapsulated cells
WO2004091763A2 (en) 2003-04-10 2004-10-28 President And Fellows Of Harvard College Formation and control of fluidic species
WO2004102204A1 (en) 2003-05-16 2004-11-25 Global Technologies (Nz) Ltd Method and apparatus for mixing sample and reagent in a suspension fluid
WO2004103565A2 (en) 2003-05-19 2004-12-02 Hans-Knöll-Institut für Naturstoff-Forschung e.V. Device and method for structuring liquids and for dosing reaction liquids into liquid compartments immersed in a separation medium
WO2004105734A1 (en) 2003-05-28 2004-12-09 Valorisation Recherche, Societe En Commandite Method of preparing microcapsules
US20040258701A1 (en) 2003-04-04 2004-12-23 Pfizer Inc. Microfluidized oil-in-water emulsions and vaccine compositions
US20050042625A1 (en) 1997-01-15 2005-02-24 Xzillion Gmbh & Co. Mass label linked hybridisation probes
WO2005021151A1 (en) 2003-08-27 2005-03-10 President And Fellows Of Harvard College Electronic control of fluidic species
WO2005023331A2 (en) 2003-09-04 2005-03-17 The United States Of America As Represented By The Department Of Veterans Affairs Hydrogel nanocompsites for ophthalmic applications
US20050079510A1 (en) 2003-01-29 2005-04-14 Jan Berka Bead emulsion nucleic acid amplification
WO2005040406A1 (en) 2003-10-17 2005-05-06 Diversa Corporation High throughput screening of antibody libraries
WO2005049787A2 (en) 2003-11-24 2005-06-02 Yeda Research And Development Co.Ltd. Compositions and methods for in vitro sorting of molecular and cellular libraries
US20050130188A1 (en) 1997-03-14 2005-06-16 The Trustees Of Tufts College Methods for detecting target analytes and enzymatic reactions
US6913935B1 (en) 1997-12-04 2005-07-05 Amersham Biosciences Uk Limited Multiple assay method
US20050181379A1 (en) 2004-02-18 2005-08-18 Intel Corporation Method and device for isolating and positioning single nucleic acid molecules
WO2005082098A2 (en) 2004-02-27 2005-09-09 President And Fellows Of Harvard College Polony fluorescent in situ sequencing beads
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US6974669B2 (en) 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US20050287572A1 (en) 2004-06-01 2005-12-29 The Regents Of The University Of California Microfabricated integrated DNA analysis system
US20060020371A1 (en) 2004-04-13 2006-01-26 President And Fellows Of Harvard College Methods and apparatus for manipulation and/or detection of biological samples and other objects
WO2006030993A1 (en) 2004-09-14 2006-03-23 Jin-Ho Choy Information code system using dna sequences
US20060073487A1 (en) 2004-10-01 2006-04-06 Oliver Kerry G System and method for inhibiting the decryption of a nucleic acid probe sequence used for the detection of a specific nucleic acid
US20060078888A1 (en) 2004-10-08 2006-04-13 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
US20060153924A1 (en) 2003-03-31 2006-07-13 Medical Research Council Selection by compartmentalised screening
WO2006078841A1 (en) 2005-01-21 2006-07-27 President And Fellows Of Harvard College Systems and methods for forming fluidic droplets encapsulated in particles such as colloidal particles
US20060199193A1 (en) 2005-03-04 2006-09-07 Tae-Woong Koo Sensor arrays and nucleic acid sequencing applications
WO2006096571A2 (en) 2005-03-04 2006-09-14 President And Fellows Of Harvard College Method and apparatus for forming multiple emulsions
US20060240506A1 (en) 2002-09-09 2006-10-26 Ariel Kushmaro Method for isolating and culturing unculturable microorganisms
JP2006289250A (en) 2005-04-08 2006-10-26 Kao Corp Micro mixer and fluid mixing method using the same
US7129091B2 (en) 2002-05-09 2006-10-31 University Of Chicago Device and method for pressure-driven plug transport and reaction
US20060257893A1 (en) 2005-02-18 2006-11-16 Toru Takahashi Devices and methods for monitoring genomic DNA of organisms
US20060263888A1 (en) 2000-06-02 2006-11-23 Honeywell International Inc. Differential white blood count on a disposable card
US20060292583A1 (en) 1999-08-30 2006-12-28 The Government of the U.S.A as represented by the Secretary of Dept. of Health and Human Services High speed parallel molecular nucleic acid sequencing
WO2007002490A2 (en) 2005-06-22 2007-01-04 The Research Foundation Of State University Of New York Massively parallel 2-dimensional capillary electrophoresis
WO2007001448A2 (en) 2004-11-04 2007-01-04 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
WO2007024840A2 (en) 2005-08-22 2007-03-01 Critical Therapeutics, Inc. Method of quantitating nucleic acids by flow cytometry microparticle-based array
US20070054119A1 (en) 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
US20070099208A1 (en) 2005-06-15 2007-05-03 Radoje Drmanac Single molecule arrays for genetic and chemical analysis
US20070154903A1 (en) 2005-06-23 2007-07-05 Nanosphere, Inc. Selective isolation and concentration of nucleic acids from complex samples
WO2007081387A1 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices, methods of use, and kits for performing diagnostics
US20070172873A1 (en) 2006-01-23 2007-07-26 Sydney Brenner Molecular counting
WO2007089541A2 (en) 2006-01-27 2007-08-09 President And Fellows Of Harvard College Fluidic droplet coalescence
US20070207060A1 (en) 2004-07-21 2007-09-06 Chengdu Kuachang Medical Industrial Limited Testing method of analytic chip of multiple reactors, the analytic chip, and the testing device
US7268167B2 (en) 2001-02-23 2007-09-11 Japan Science And Technology Agency Process for producing emulsion and microcapsules and apparatus therefor
US20070228588A1 (en) 2006-03-30 2007-10-04 Yasuko Noritomi Apparatus for producing particles, emulsifier holding member, method for producing particles, and method for producing molecular membrane
WO2007114794A1 (en) 2006-03-31 2007-10-11 Nam Trung Nguyen Active control for droplet-based microfluidics
WO2007121489A2 (en) 2006-04-19 2007-10-25 Applera Corporation Reagents, methods, and libraries for gel-free bead-based sequencing
US20070264320A1 (en) 2006-05-09 2007-11-15 The Regents Of The University Of California Microfluidic device for forming monodisperse lipoplexes
WO2007133710A2 (en) 2006-05-11 2007-11-22 Raindance Technologies, Inc. Microfluidic devices and methods of use thereof
WO2007138178A2 (en) 2006-05-30 2007-12-06 Centre National De La Recherche Scientifique Method for treating drops in a microfluid circuit
WO2007139766A2 (en) 2006-05-22 2007-12-06 Nanostring Technologies, Inc. Systems and methods for analyzing nanoreporters
WO2007140015A2 (en) 2006-05-26 2007-12-06 Althea Technologies, Inc Biochemical analysis of partitioned cells
WO2007149432A2 (en) 2006-06-19 2007-12-27 The Johns Hopkins University Single-molecule pcr on microparticles in water-in-oil emulsions
US20080004436A1 (en) 2004-11-15 2008-01-03 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Directed Evolution and Selection Using in Vitro Compartmentalization
WO2008021123A1 (en) 2006-08-07 2008-02-21 President And Fellows Of Harvard College Fluorocarbon emulsion stabilizing surfactants
WO2008091792A2 (en) 2007-01-23 2008-07-31 Honeywell International Inc. Hydrogel microarray with embedded metal nanoparticles
WO2008102057A1 (en) 2007-02-21 2008-08-28 Valtion Teknillinen Tutkimuskeskus Method and test kit for determining the amounts of target sequences and nucleotide variations therein
WO2008109176A2 (en) 2007-03-07 2008-09-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
US20080241820A1 (en) 2007-02-16 2008-10-02 Krutzik Peter O Multiplex cellular assays using detectable cell barcodes
WO2008121342A2 (en) 2007-03-28 2008-10-09 President And Fellows Of Harvard College Emulsions and techniques for formation
WO2008134153A1 (en) 2007-04-23 2008-11-06 Advanced Liquid Logic, Inc. Bead-based multiplexed analytical methods and instrumentation
US20090005252A1 (en) 2006-02-24 2009-01-01 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20090011943A1 (en) 2005-06-15 2009-01-08 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
WO2009005680A1 (en) 2007-06-29 2009-01-08 President And Fellows Of Harvard College Methods and apparatus for manipulation of fluidic species
WO2009011808A1 (en) 2007-07-13 2009-01-22 President And Fellows Of Harvard College Droplet-based selection
US20090025277A1 (en) 2005-02-21 2009-01-29 Kagoshima University Method for purifying biodiesel fuel
US20090035770A1 (en) 2006-10-25 2009-02-05 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
US20090053169A1 (en) 2007-08-20 2009-02-26 Pharmain Corporation Oligonucleotide Core Carrier Compositions for Delivery of Nucleic Acid-Containing Therapeutic Agents, Methods of Making and Using the Same
US20090098555A1 (en) 2007-09-26 2009-04-16 President And Fellows Of Harvard College Methods and applications for stitched dna barcodes
WO2009061372A1 (en) 2007-11-02 2009-05-14 President And Fellows Of Harvard College Systems and methods for creating multi-phase entities, including particles and/or fluids
US7536928B2 (en) 2005-06-16 2009-05-26 Ntn Corporation Ball screw
WO2009085215A1 (en) 2007-12-21 2009-07-09 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
US20090197772A1 (en) 2004-03-31 2009-08-06 Andrew Griffiths Compartmentalised combinatorial chemistry by microfluidic control
US20090202984A1 (en) 2008-01-17 2009-08-13 Sequenom, Inc. Single molecule nucleic acid sequence analysis processes and compositions
JP2009208074A (en) 2008-02-08 2009-09-17 Kao Corp Manufacturing method of fine particle dispersion liquid
US20090286687A1 (en) 2003-07-05 2009-11-19 The Johns Hopkins University Method and Compositions for Detection and Enumeration of Genetic Variations
US7622280B2 (en) 2001-11-16 2009-11-24 454 Life Sciences Corporation Emulsion compositions
US7645596B2 (en) 1998-05-01 2010-01-12 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
WO2010004018A2 (en) 2008-07-11 2010-01-14 Eth Zurich Degradable microcapsules
US20100022414A1 (en) 2008-07-18 2010-01-28 Raindance Technologies, Inc. Droplet Libraries
US20100021984A1 (en) 2008-05-23 2010-01-28 Edd Jon F Microfluidic Droplet Encapsulation
US20100021973A1 (en) * 2005-08-02 2010-01-28 Makarov Vladimir L Compositions and methods for processing and amplification of dna, including using multiple enzymes in a single reaction
US7666664B2 (en) 2006-07-14 2010-02-23 Roche Molecular Systems, Inc. Instrument for heating and cooling
US20100069263A1 (en) 2008-09-12 2010-03-18 Washington, University Of Sequence tag directed subassembly of short sequencing reads into long sequencing reads
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
EP1967592B1 (en) 1995-06-07 2010-04-28 Solexa, Inc. Method of improving the efficiency of polynucleotide sequencing
US7745178B2 (en) 1999-10-27 2010-06-29 Affymetrix, Inc. Complexity management of genomic DNA
US20100173394A1 (en) 2008-09-23 2010-07-08 Colston Jr Billy Wayne Droplet-based assay system
US20100210479A1 (en) 2003-03-31 2010-08-19 Medical Research Council Method of synthesis and testing of cominatorial libraries using microcapsules
WO2010148039A2 (en) 2009-06-15 2010-12-23 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
WO2010151776A2 (en) 2009-06-26 2010-12-29 President And Fellows Of Harvard College Fluid injection
US20110033854A1 (en) 2007-12-05 2011-02-10 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
US7910354B2 (en) 2006-10-27 2011-03-22 Complete Genomics, Inc. Efficient arrays of amplified polynucleotides
WO2011047870A1 (en) 2009-10-22 2011-04-28 Plasticell Ltd Nested cell encapsulation
WO2011056546A1 (en) 2009-10-27 2011-05-12 President And Fellows Of Harvard College Droplet creation techniques
WO2011066476A1 (en) 2009-11-25 2011-06-03 Quantalife, Inc. Methods and compositions for detecting genetic material
US7960104B2 (en) 2005-10-07 2011-06-14 Callida Genomics, Inc. Self-assembled single molecule arrays and uses thereof
US20110160078A1 (en) 2009-12-15 2011-06-30 Affymetrix, Inc. Digital Counting of Individual Molecules by Stochastic Attachment of Diverse Labels
US20110195496A1 (en) 2003-09-25 2011-08-11 Atsushi Muraguchi Microwell array chip and method of manufacturing same
US20110217736A1 (en) 2010-03-02 2011-09-08 Quantalife, Inc. System for hot-start amplification via a multiple emulsion
US20110305761A1 (en) 2008-06-05 2011-12-15 President And Fellows Of Harvard College Polymersomes, colloidosomes, liposomes, and other species associated with fluidic droplets
US20120000777A1 (en) 2010-06-04 2012-01-05 The Regents Of The University Of California Devices and methods for forming double emulsion droplet compositions and polymer particles
US20120015822A1 (en) 2008-12-19 2012-01-19 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
WO2012012037A1 (en) * 2010-07-19 2012-01-26 New England Biolabs, Inc. Oligonucleotide adaptors: compositions and methods of use
US20120071331A1 (en) 2010-09-21 2012-03-22 James Casbon Increasing confidence of allele calls with molecular counting
WO2012048341A1 (en) 2010-10-08 2012-04-12 President And Fellows Of Harvard College High-throughput single cell barcoding
US20120121481A1 (en) 2009-03-13 2012-05-17 President And Fellows Of Harvard College Scale-up of flow-focusing microfluidic devices
GB2485850A (en) 2009-11-25 2012-05-30 Bio Rad Laboratories DNA copy number and Chromosome aneuploidy detection by amplification wherein the ligated products are partitioned into oil droplets prior to amplification
WO2012083225A2 (en) 2010-12-16 2012-06-21 Gigagen, Inc. System and methods for massively parallel analysis of nycleic acids in single cells
US20120172259A1 (en) 2008-07-02 2012-07-05 Illumina Cambridge Limited Using populations of beads for the fabrication of arrays on surfaces
US20120190032A1 (en) 2010-03-25 2012-07-26 Ness Kevin D Droplet generation for droplet-based assays
US20120196288A1 (en) * 2011-01-27 2012-08-02 Lawrence Livermore National Security, Llc Chip-Based Droplet Sorting
EP1905828B1 (en) 1999-01-07 2012-08-08 Medical Research Council Optical sorting method
US20120211084A1 (en) 2009-09-02 2012-08-23 President And Fellows Of Harvard College Multiple emulsions created using jetting and other techniques
US8252539B2 (en) 2000-09-15 2012-08-28 California Institute Of Technology Microfabricated crossflow devices and methods
US20120220497A1 (en) 2009-11-03 2012-08-30 Gen 9, Inc. Methods and Microfluidic Devices for the Manipulation of Droplets in High Fidelity Polynucleotide Assembly
US20120220494A1 (en) 2011-02-18 2012-08-30 Raindance Technolgies, Inc. Compositions and methods for molecular labeling
US8273573B2 (en) 2002-05-09 2012-09-25 The University Of Chicago Method for obtaining a collection of plugs comprising biological molecules
WO2012149042A2 (en) 2011-04-25 2012-11-01 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
WO2012166425A2 (en) 2011-05-27 2012-12-06 President And Fellows Of Harvard College Methods of amplifying whole genome of a single cell
US20120309002A1 (en) 2011-06-02 2012-12-06 Raindance Technologies, Inc. Sample multiplexing
US20130028812A1 (en) 2010-10-07 2013-01-31 The Regents Of The University Of California Methods and systems for on demand droplet generation and impedance based detection
US20130046030A1 (en) 2011-05-23 2013-02-21 Basf Se Control of emulsions, including multiple emulsions
US20130079231A1 (en) 2011-09-09 2013-03-28 The Board Of Trustees Of The Leland Stanford Junior University Methods for obtaining a sequence
US20130109575A1 (en) 2009-12-23 2013-05-02 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
US20130157899A1 (en) 2007-12-05 2013-06-20 Perkinelmer Health Sciences, Inc. Reagents and methods relating to dna assays using amplicon probes on encoded particles
US20130189700A1 (en) 2011-07-25 2013-07-25 Bio-Rad Laboratories, Inc. Breakage of an emulsion containing nucleic acid
US20130225418A1 (en) 2012-02-24 2013-08-29 Andrew Watson Labeling and sample preparation for sequencing
WO2013177220A1 (en) 2012-05-21 2013-11-28 The Scripps Research Institute Methods of sample preparation
US8603749B2 (en) 2006-11-15 2013-12-10 Biospherex, LLC Multitag sequencing ecogenomics analysis-US
WO2014028537A1 (en) 2012-08-14 2014-02-20 10X Technologies, Inc. Microcapsule compositions and methods
US20140206554A1 (en) 2012-12-14 2014-07-24 10X Technologies, Inc. Methods and Systems for Processing Polynucleotides
US20140227684A1 (en) 2013-02-08 2014-08-14 10X Technologies, Inc. Partitioning and processing of analytes and other species
US20140227706A1 (en) 2011-05-16 2014-08-14 Dna Chip Research Inc. Method for assessing progression of clinical state of malignant neoplasm by quantitative detection of DNA in blood
US20140378349A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20140378322A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20140378350A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20150005199A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
US20150005200A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
US20150111256A1 (en) 2012-02-17 2015-04-23 President And Fellows Of Harvard College Assembly of Nucleic Acid Sequences in Emulsions
US20150292988A1 (en) 2014-04-10 2015-10-15 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same

Family Cites Families (570)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1007137A (en) 1908-06-08 1911-10-31 Theodore D Palmer Cloth-pressing machine.
US1004111A (en) 1908-10-17 1911-09-26 Westinghouse Air Brake Co Fluid-pressure brake.
US1003026A (en) 1911-04-12 1911-09-12 Bernard Bartmann Bobbin-holder.
US1005998A (en) 1911-07-10 1911-10-17 Arthur Munchausen Berry-carrier.
US1005372A (en) 1911-08-11 1911-10-10 Albert Gay Plow-fender.
US2797149A (en) 1953-01-08 1957-06-25 Technicon International Ltd Methods of and apparatus for analyzing liquids containing crystalloid and non-crystalloid constituents
US3047367A (en) 1959-12-01 1962-07-31 Technicon Instr Automatic analysis with fluid segmentation
US3479141A (en) 1967-05-17 1969-11-18 Technicon Corp Method and apparatus for analysis
US4124638A (en) 1977-09-12 1978-11-07 Hansen John N Solubilizable polyacrylamide gels containing disulfide cross-linkages
US4253846A (en) 1979-11-21 1981-03-03 Technicon Instruments Corporation Method and apparatus for automated analysis of fluid samples
GB2097692B (en) 1981-01-10 1985-05-22 Shaw Stewart P D Combining chemical reagents
DE3230289A1 (en) 1982-08-14 1984-02-16 Bayer Ag, 5090 Leverkusen PRODUCTION OF PHARMACEUTICAL OR COSMETIC DISPERSIONS
US4582802A (en) 1983-09-30 1986-04-15 The United States Of America As Represented By The Department Of Health And Human Services Stimulation of enzymatic ligation of DNA by high concentrations of nonspecific polymers
JPS60227826A (en) 1984-04-27 1985-11-13 Sogo Yatsukou Kk Graft capsule responding to ph
KR890003947B1 (en) * 1985-12-11 1989-10-13 가부시기가이샤 시마즈세이사구쇼 Apparatus for cell fusion
US4872895A (en) 1986-12-11 1989-10-10 American Telephone And Telegraph Company, At&T Bell Laboratories Method for fabricating articles which include high silica glass bodies
US5137829A (en) 1987-10-05 1992-08-11 Washington University DNA transposon TN5SEQ1
US5185099A (en) 1988-04-20 1993-02-09 Institut National De Recherche Chimique Appliquee Visco-elastic, isotropic materials based on water, fluorinate sufactants and fluorinated oils, process for their preparation, and their use in various fields, such as optics, pharmacology and electrodynamics
US5237016A (en) 1989-01-05 1993-08-17 Siska Diagnostics, Inc. End-attachment of oligonucleotides to polyacrylamide solid supports for capture and detection of nucleic acids
US6176962B1 (en) 1990-02-28 2001-01-23 Aclara Biosciences, Inc. Methods for fabricating enclosed microchannel structures
US5756334A (en) 1990-04-26 1998-05-26 New England Biolabs, Inc. Thermostable DNA polymerase from 9°N-7 and methods for producing the same
JP3392863B2 (en) 1990-07-24 2003-03-31 エフ.ホフマン ― ラ ロシュ アーゲー Reduction of non-specific amplification in in vitro nucleic acid amplification using modified nucleobases
US5489523A (en) 1990-12-03 1996-02-06 Stratagene Exonuclease-deficient thermostable Pyrococcus furiosus DNA polymerase I
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5270183A (en) 1991-02-08 1993-12-14 Beckman Research Institute Of The City Of Hope Device and method for the automated cycling of solutions between two or more temperatures
EP0594772B1 (en) 1991-07-04 1996-08-28 Immunodex K/S Water-soluble, polymer-based reagents and conjugates comprising moieties derived from divinyl sulfone
JPH06509473A (en) 1991-08-10 1994-10-27 メディカル・リサーチ・カウンシル Processing of cell populations
DE69217497T2 (en) 1991-09-18 1997-06-12 Affymax Tech Nv METHOD FOR SYNTHESISING THE DIFFERENT COLLECTIONS OF OLIGOMERS
US5840865A (en) 1992-09-14 1998-11-24 Institute Of Molecular Biology And Biotechnology/Forth Eukaryotic transposable element
US5897783A (en) 1992-09-24 1999-04-27 Amersham International Plc Magnetic separation method
US5569364A (en) 1992-11-05 1996-10-29 Soane Biosciences, Inc. Separation media for electrophoresis
IL108497A0 (en) 1993-02-01 1994-05-30 Seq Ltd Methods and apparatus for dna sequencing
WO1994019101A1 (en) 1993-02-16 1994-09-01 Alliance Pharmaceutical Corp. Method of microemulsifying fluorinated oils
AU6637394A (en) 1993-04-19 1994-11-08 Medisorb Technologies International L.P. Long-acting treatment by slow-release delivery of antisense oligodeoxyribonucleotides from biodegradable microparticles
US5456986A (en) * 1993-06-30 1995-10-10 Carnegie Mellon University Magnetic metal or metal carbide nanoparticles and a process for forming same
EP0636413B1 (en) 1993-07-28 2001-11-14 PE Corporation (NY) Nucleic acid amplification reaction apparatus and method
US5874239A (en) 1993-07-30 1999-02-23 Affymax Technologies N.V. Biotinylation of proteins
DE69433425T2 (en) 1993-08-30 2004-10-07 Promega Corp COMPOSITIONS AND METHOD FOR PURIFYING NUCLEIC ACIDS
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5558071A (en) 1994-03-07 1996-09-24 Combustion Electromagnetics, Inc. Ignition system power converter and controller
US5648211A (en) 1994-04-18 1997-07-15 Becton, Dickinson And Company Strand displacement amplification using thermophilic enzymes
US5705628A (en) 1994-09-20 1998-01-06 Whitehead Institute For Biomedical Research DNA purification and isolation using magnetic particles
US5585069A (en) 1994-11-10 1996-12-17 David Sarnoff Research Center, Inc. Partitioned microelectronic and fluidic device array for clinical diagnostics and chemical synthesis
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
US6866760B2 (en) 1998-08-27 2005-03-15 E Ink Corporation Electrophoretic medium and process for the production thereof
US5872010A (en) 1995-07-21 1999-02-16 Northeastern University Microscale fluid handling system
US6057149A (en) 1995-09-15 2000-05-02 The University Of Michigan Microscale devices and reactions in microscale devices
WO1997039359A1 (en) 1996-04-15 1997-10-23 Dade International Inc. Apparatus and method for analysis
EP0912761A4 (en) 1996-05-29 2004-06-09 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US5846727A (en) 1996-06-06 1998-12-08 Board Of Supervisors Of Louisiana State University And Agricultural & Mechanical College Microsystem for rapid DNA sequencing
AU4114397A (en) 1996-07-15 1998-02-09 Kemgas Limited Production of powders
US5965443A (en) 1996-09-09 1999-10-12 Wisconsin Alumni Research Foundation System for in vitro transposition
US5900481A (en) 1996-11-06 1999-05-04 Sequenom, Inc. Bead linkers for immobilizing nucleic acids to solid supports
US6133436A (en) 1996-11-06 2000-10-17 Sequenom, Inc. Beads bound to a solid support and to nucleic acids
US6379929B1 (en) 1996-11-20 2002-04-30 The Regents Of The University Of Michigan Chip-based isothermal amplification devices and methods
US20020172965A1 (en) 1996-12-13 2002-11-21 Arcaris, Inc. Methods for measuring relative amounts of nucleic acids in a complex mixture and retrieval of specific sequences therefrom
DE69825601T2 (en) 1997-02-12 2005-04-28 Chan, Eugene Y, Brookline METHOD FOR THE ANALYSIS OF POLYMERS
US20030027203A1 (en) 1997-03-24 2003-02-06 Fields Robert E. Biomolecular processor
EP0975807B1 (en) 1997-05-02 2006-09-27 Gen-Probe Incorporated Two-step hybridization and capture of a polynucleotide
AU734957B2 (en) 1997-05-16 2001-06-28 Alberta Research Council Inc. Microfluidic system and methods of use
FI103809B1 (en) 1997-07-14 1999-09-30 Finnzymes Oy In vitro method for producing templates for DNA sequencing
US20050037397A1 (en) 2001-03-28 2005-02-17 Nanosphere, Inc. Bio-barcode based detection of target analytes
US6368871B1 (en) 1997-08-13 2002-04-09 Cepheid Non-planar microstructures for manipulation of fluid samples
US7214298B2 (en) 1997-09-23 2007-05-08 California Institute Of Technology Microfabricated cell sorter
US5842787A (en) 1997-10-09 1998-12-01 Caliper Technologies Corporation Microfluidic systems incorporating varied channel dimensions
JP2001521169A (en) 1997-10-24 2001-11-06 ノースイースタン・ユニバーシティ Multi-channel micro-scale system for high-throughput coordinated separation with extensive collection and analysis
WO1999033963A1 (en) 1997-12-31 1999-07-08 Chiron Corporation Metastatic cancer regulated gene
CA2321262A1 (en) 1998-02-19 1999-08-26 President And Fellows Of Harvard College Monovalent, multivalent, and multimeric mhc binding domain fusion proteins and conjugates, and uses therefor
WO1999050402A1 (en) 1998-03-27 1999-10-07 President And Fellows Of Harvard College Systematic identification of essential genes by in vitro transposon mutagenesis
US6022716A (en) 1998-04-10 2000-02-08 Genset Sa High throughput DNA sequencing vector
US5997636A (en) 1998-05-01 1999-12-07 Instrumentation Technology Associates, Inc. Method and apparatus for growing crystals
US6123798A (en) 1998-05-06 2000-09-26 Caliper Technologies Corp. Methods of fabricating polymeric structures incorporating microscale fluidic elements
US6534262B1 (en) 1998-05-14 2003-03-18 Whitehead Institute For Biomedical Research Solid phase technique for selectively isolating nucleic acids
US6306590B1 (en) 1998-06-08 2001-10-23 Caliper Technologies Corp. Microfluidic matrix localization apparatus and methods
EP2045334A1 (en) 1998-06-24 2009-04-08 Illumina, Inc. Decoding of array sensors with microspheres
US6159736A (en) 1998-09-23 2000-12-12 Wisconsin Alumni Research Foundation Method for making insertional mutations using a Tn5 synaptic complex
AR021833A1 (en) 1998-09-30 2002-08-07 Applied Research Systems METHODS OF AMPLIFICATION AND SEQUENCING OF NUCLEIC ACID
AU763497B2 (en) 1998-10-13 2003-07-24 Biomicro Systems, Inc. Fluid circuit components based upon passive fluid dynamics
SE9803614L (en) 1998-10-19 2000-04-20 Muhammed Mamoun Method and apparatus for producing nanoparticles
US6569631B1 (en) 1998-11-12 2003-05-27 3-Dimensional Pharmaceuticals, Inc. Microplate thermal shift assay for ligand development using 5-(4″dimethylaminophenyl)-2-(4′-phenyl)oxazole derivative fluorescent dyes
US5942609A (en) 1998-11-12 1999-08-24 The Porkin-Elmer Corporation Ligation assembly and detection of polynucleotides on solid-support
WO2000034527A2 (en) 1998-12-11 2000-06-15 The Regents Of The University Of California Targeted molecular bar codes
NO986133D0 (en) 1998-12-23 1998-12-23 Preben Lexow Method of DNA Sequencing
US6416642B1 (en) 1999-01-21 2002-07-09 Caliper Technologies Corp. Method and apparatus for continuous liquid flow in microscale channels using pressure injection, wicking, and electrokinetic injection
US20030027214A1 (en) 1999-02-17 2003-02-06 Kamb Carl Alexander Methods for substrate-ligand interaction screening
US7615373B2 (en) 1999-02-25 2009-11-10 Virginia Commonwealth University Intellectual Property Foundation Electroprocessed collagen and tissue engineering
US6171850B1 (en) 1999-03-08 2001-01-09 Caliper Technologies Corp. Integrated devices and systems for performing temperature controlled reactions and analyses
US6303343B1 (en) 1999-04-06 2001-10-16 Caliper Technologies Corp. Inefficient fast PCR
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
ATE413467T1 (en) 1999-04-20 2008-11-15 Illumina Inc DETECTION OF NUCLEIC ACID REACTIONS ON BEAD ARRAYS
JP2002542781A (en) 1999-04-28 2002-12-17 ザ、ボード、オブ、トラスティーズ、オブ、ザ、リーランド、スタンフォード、ジュニア、ユニバーシティ P factor-derived vectors and methods of use
WO2000070095A2 (en) 1999-05-17 2000-11-23 Dade Behring Inc. Homogeneous isothermal amplification and detection of nucleic acids using a template switch oligonucleotide
US20020051971A1 (en) 1999-05-21 2002-05-02 John R. Stuelpnagel Use of microfluidic systems in the detection of target analytes using microsphere arrays
US6846622B1 (en) 1999-05-26 2005-01-25 Oregon Health & Science University Tagged epitope protein transposable element
US20030124509A1 (en) 1999-06-03 2003-07-03 Kenis Paul J.A. Laminar flow patterning and articles made thereby
AU6068300A (en) 1999-07-06 2001-01-22 Caliper Technologies Corporation Microfluidic systems and methods for determining modulator kinetics
US6977145B2 (en) 1999-07-28 2005-12-20 Serono Genetics Institute S.A. Method for carrying out a biochemical protocol in continuous flow in a microreactor
EP1210358A4 (en) 1999-08-13 2005-01-05 Univ Brandeis Detection of nucleic acids
EP1208238B1 (en) 1999-08-27 2008-11-12 Matrix Technologies Corporation Methods of immobilizing ligands on solid supports
WO2001027610A2 (en) 1999-10-13 2001-04-19 Signature Bioscience, Inc. System and method for detecting and identifying molecular events in a test sample
AU1100201A (en) 1999-10-28 2001-05-08 Board Of Trustees Of The Leland Stanford Junior University Methods of in vivo gene transfer using a sleeping beauty transposon system
JP4721603B2 (en) 1999-11-08 2011-07-13 栄研化学株式会社 Mutation and / or polymorphism detection method
US6432290B1 (en) 1999-11-26 2002-08-13 The Governors Of The University Of Alberta Apparatus and method for trapping bead based reagents within microfluidic analysis systems
US20010051348A1 (en) 2000-01-28 2001-12-13 Lee Chee Wee Novel ligands and methods for preparing same
CA2401118A1 (en) 2000-02-23 2001-08-30 Zyomyx, Inc. Microfluidic devices and methods
CA2399199A1 (en) 2000-02-23 2001-08-30 Ring-Ling Chien Multi-reservoir pressure control system
IL134830A0 (en) 2000-03-01 2001-05-20 Chay 13 Medical Res Group N V Peptides and immunostimulatory and anti-bacterial pharmaceutical compositions containing them
AU2001252201A1 (en) 2000-03-14 2001-09-24 Amylin Pharmaceuticals, Inc. Effects of glucagon-like peptide-1 (7-36) on antro-pyloro-duodenal motility
US6409832B2 (en) 2000-03-31 2002-06-25 Micronics, Inc. Protein crystallization in microfluidic structures
US20020001856A1 (en) 2000-04-06 2002-01-03 Chow Andrea W. Methods and devices for achieving long incubation times in high-throughput systems
DK2278030T3 (en) 2000-04-10 2017-07-24 Taxon Biosciences Inc METHODS OF RESEARCH AND GENETIC ANALYSIS OF POPULATIONS
US6481453B1 (en) 2000-04-14 2002-11-19 Nanostream, Inc. Microfluidic branch metering systems and methods
US20060008799A1 (en) 2000-05-22 2006-01-12 Hong Cai Rapid haplotyping by single molecule detection
US20010042712A1 (en) 2000-05-24 2001-11-22 Battrell C. Frederick Microfluidic concentration gradient loop
AU2001281076A1 (en) 2000-08-07 2002-02-18 Nanostream, Inc. Fluidic mixer in microfluidic system
US6773566B2 (en) 2000-08-31 2004-08-10 Nanolytics, Inc. Electrostatic actuators for microfluidics and methods for using same
US6610499B1 (en) 2000-08-31 2003-08-26 The Regents Of The University Of California Capillary array and related methods
EP1317569B1 (en) 2000-09-14 2009-11-18 Caliper Life Sciences, Inc. Microfluidic devices and methods for performing temperature mediated reactions
US7258774B2 (en) 2000-10-03 2007-08-21 California Institute Of Technology Microfluidic devices and methods of use
US6492154B2 (en) 2001-01-31 2002-12-10 Applera Corporation Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
CA2332186A1 (en) 2001-02-08 2002-08-08 Her Majesty In Right Of Canada As Represented By The Minister Of Agricul Ture And Agri-Food Canada Replicative in vivo gene targeting
US7670559B2 (en) 2001-02-15 2010-03-02 Caliper Life Sciences, Inc. Microfluidic systems with enhanced detection systems
EP1368309B2 (en) 2001-02-22 2014-10-01 Anika Therapeutics Inc. Thiol-modified hyaluronan
US20150329617A1 (en) 2001-03-14 2015-11-19 Dynal Biotech Asa Novel MHC molecule constructs, and methods of employing these constructs for diagnosis and therapy, and uses of MHC molecules
US7211654B2 (en) 2001-03-14 2007-05-01 Regents Of The University Of Michigan Linkers and co-coupling agents for optimization of oligonucleotide synthesis and purification on solid supports
WO2002082057A2 (en) 2001-04-03 2002-10-17 Micronics, Inc. Split focusing cytometer
US7138267B1 (en) 2001-04-04 2006-11-21 Epicentre Technologies Corporation Methods and compositions for amplifying DNA clone copy number
US20030027221A1 (en) * 2001-04-06 2003-02-06 Scott Melissa E. High-throughput screening assays by encapsulation
US6880576B2 (en) 2001-06-07 2005-04-19 Nanostream, Inc. Microfluidic devices for methods development
US7179423B2 (en) 2001-06-20 2007-02-20 Cytonome, Inc. Microfluidic system including a virtual wall fluid interface port for interfacing fluids with the microfluidic system
US7262063B2 (en) 2001-06-21 2007-08-28 Bio Array Solutions, Ltd. Directed assembly of functional heterostructures
US7682353B2 (en) 2001-06-29 2010-03-23 Coloplast A/S Catheter device
US7077152B2 (en) 2001-07-07 2006-07-18 Nanostream, Inc. Microfluidic metering systems and methods
WO2003008437A2 (en) 2001-07-20 2003-01-30 California Institute Of Technology Protein and nucleic acid expression systems
US20030148335A1 (en) 2001-10-10 2003-08-07 Li Shen Detecting targets by unique identifier nucleotide tags
US7297485B2 (en) 2001-10-15 2007-11-20 Qiagen Gmbh Method for nucleic acid amplification that results in low amplification bias
WO2003044221A1 (en) 2001-10-19 2003-05-30 West Virginia University Research Corporation Microfluidic system for proteome analysis
US6783647B2 (en) 2001-10-19 2004-08-31 Ut-Battelle, Llc Microfluidic systems and methods of transport and lysis of cells and analysis of cell lysate
US20030149307A1 (en) 2001-10-24 2003-08-07 Baxter International Inc. Process for the preparation of polyethylene glycol bis amine
US20040195728A1 (en) 2001-10-26 2004-10-07 Dennis Slomski System and method for injection molded micro-replication of micro-fluidic substrates
US7262056B2 (en) 2001-11-08 2007-08-28 Mirus Bio Corporation Enhancing intermolecular integration of nucleic acids using integrator complexes
ATE458832T1 (en) 2001-11-28 2010-03-15 Bio Rad Laboratories PARALLEL SCORING OF POLYMORPHISMS USING AMPLIFICATION AND ERROR CORRECTION
US7335153B2 (en) 2001-12-28 2008-02-26 Bio Array Solutions Ltd. Arrays of microparticles and methods of preparation thereof
AU2003202026A1 (en) 2002-01-16 2003-09-02 Dynal Biotech Asa Method for isolating nucleic acids and protein from a single sample
KR100459870B1 (en) 2002-02-22 2004-12-04 한국과학기술원 CONSTRUCTION OF NOVEL STRAINS CONTAINING MINIMIZING GENOME BY Tn5-COUPLED Cre/loxP EXCISION SYSTEM
EP1488006B1 (en) * 2002-03-20 2008-05-28 InnovativeBio.Biz Microcapsules with controlable permeability encapsulating a nucleic acid amplification reaction mixture and their use as reaction compartments for parallels reactions
US7527966B2 (en) 2002-06-26 2009-05-05 Transgenrx, Inc. Gene regulation in transgenic animals using a transposon-based vector
EP1546376B9 (en) 2002-09-30 2009-08-12 F.Hoffmann-La Roche Ag Oligonucleotides for genotyping thymidylate synthase gene
US20040081962A1 (en) 2002-10-23 2004-04-29 Caifu Chen Methods for synthesizing complementary DNA
US6979713B2 (en) 2002-11-25 2005-12-27 3M Innovative Properties Company Curable compositions and abrasive articles therefrom
US20050266582A1 (en) 2002-12-16 2005-12-01 Modlin Douglas N Microfluidic system with integrated permeable membrane
US20040248299A1 (en) 2002-12-27 2004-12-09 Sumedha Jayasena RNA interference
CA2513302C (en) 2003-01-17 2013-03-26 The Trustees Of Boston University Haplotype analysis
DK1594973T3 (en) 2003-02-10 2012-02-13 Max Delbrueck Centrum Transposon-based targeting system
US10533998B2 (en) 2008-07-18 2020-01-14 Bio-Rad Laboratories, Inc. Enzyme quantification
US7316903B2 (en) 2003-03-28 2008-01-08 United States Of America As Represented By The Department Of Health And Human Services Detection of nucleic acid sequence variations using phase Mu transposase
US20100035254A1 (en) 2003-04-08 2010-02-11 Pacific Biosciences Of California, Inc. Composition and method for nucleic acid sequencing
GB0313170D0 (en) 2003-06-09 2003-07-16 Qinetiq Ltd Method and apparatus for spore disruption and/or detection
WO2004113877A1 (en) 2003-06-13 2004-12-29 The General Hospital Corporation Microfluidic systems for size based removal of red blood cells and platelets from blood
GB2403475B (en) 2003-07-01 2008-02-06 Oxitec Ltd Stable integrands
GB0315438D0 (en) 2003-07-02 2003-08-06 Univ Manchester Analysis of mixed cell populations
WO2005023427A1 (en) 2003-09-05 2005-03-17 Stokes Bio Limited A microfluidic analysis system
US7354706B2 (en) 2003-09-09 2008-04-08 The Regents Of The University Of Colorado, A Body Corporate Use of photopolymerization for amplification and detection of a molecular recognition event
EP1672064A4 (en) 2003-09-22 2006-12-20 Riken Efficient method of preparing dna inverted repeat structure
EP1694869A2 (en) 2003-11-10 2006-08-30 Investigen, Inc. Methods of preparing nucleic acid for detection
US20050136417A1 (en) 2003-12-19 2005-06-23 Affymetrix, Inc. Amplification of nucleic acids
US8071364B2 (en) 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
US7927797B2 (en) 2004-01-28 2011-04-19 454 Life Sciences Corporation Nucleic acid amplification with continuous flow emulsion
US20100216153A1 (en) 2004-02-27 2010-08-26 Helicos Biosciences Corporation Methods for detecting fetal nucleic acids and diagnosing fetal abnormalities
KR100552706B1 (en) 2004-03-12 2006-02-20 삼성전자주식회사 Method and apparatus for nucleic acid amplification
CN100431679C (en) 2004-03-23 2008-11-12 独立行政法人科学技术振兴机构 Method and device for producing micro-droplets
WO2005111242A2 (en) 2004-05-10 2005-11-24 Parallele Bioscience, Inc. Digital profiling of polynucleotide populations
US20080268507A1 (en) 2004-05-25 2008-10-30 Airbus Deutschland Gmbh Recombinant Dna Nicking Endonuclease and Uses Thereof
US7700281B2 (en) 2004-06-30 2010-04-20 Usb Corporation Hot start nucleic acid amplification
US7968085B2 (en) 2004-07-05 2011-06-28 Ascendis Pharma A/S Hydrogel formulations
US7608434B2 (en) 2004-08-04 2009-10-27 Wisconsin Alumni Research Foundation Mutated Tn5 transposase proteins and the use thereof
US7329493B2 (en) 2004-12-22 2008-02-12 Asiagen Corporation One-tube nested PCR for detecting Mycobacterium tuberculosis
EP1871527B1 (en) 2004-12-23 2017-09-27 Abbott Point of Care Inc. Molecular diagnostics system
US7579153B2 (en) 2005-01-25 2009-08-25 Population Genetics Technologies, Ltd. Isothermal DNA amplification
US7407757B2 (en) 2005-02-10 2008-08-05 Population Genetics Technologies Genetic analysis by sequence-specific sorting
US7393665B2 (en) 2005-02-10 2008-07-01 Population Genetics Technologies Ltd Methods and compositions for tagging and identifying polynucleotides
US8407013B2 (en) 2005-06-07 2013-03-26 Peter K. Rogan AB initio generation of single copy genomic probes
US9175295B2 (en) 2005-07-05 2015-11-03 The Chemo-Sero-Therapeutic Research Institute Modified transposon vector and its use
JP5051490B2 (en) 2005-07-08 2012-10-17 独立行政法人産業技術総合研究所 Inorganic microcapsule encapsulating macro-biomaterial and method for producing the same
US20070020640A1 (en) 2005-07-21 2007-01-25 Mccloskey Megan L Molecular encoding of nucleic acid templates for PCR and other forms of sequence analysis
FR2888912B1 (en) 2005-07-25 2007-08-24 Commissariat Energie Atomique METHOD FOR CONTROLLING COMMUNICATION BETWEEN TWO ZONES BY ELECTROWRINKING, DEVICE COMPRISING ISOLABLE ZONES AND OTHERS AND METHOD FOR PRODUCING SUCH DEVICE
DE102005037401B4 (en) 2005-08-08 2007-09-27 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Formation of an emulsion in a fluidic microsystem
US7556776B2 (en) 2005-09-08 2009-07-07 President And Fellows Of Harvard College Microfluidic manipulation of fluids and reactions
JP2007074967A (en) 2005-09-13 2007-03-29 Canon Inc Identifier probe and method for amplifying nucleic acid by using the same
JP2009513948A (en) 2005-09-16 2009-04-02 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Colorimetric bio barcode amplification assay for analyte detection
US20070111241A1 (en) 2005-10-14 2007-05-17 Nezih Cereb System and method for accessing, tracking, and editing sequence analysis and software to accomplish the same
US7709544B2 (en) 2005-10-25 2010-05-04 Massachusetts Institute Of Technology Microstructure synthesis by flow lithography and polymerization
WO2007120265A2 (en) * 2005-11-14 2007-10-25 Applera Corporation Coded molecules for detecting target analytes
US20070134277A1 (en) 2005-12-09 2007-06-14 Children's Medical Center Corporation Pharmaceutical formulation for sulfur-containing drugs in liquid dosage forms
US20070141584A1 (en) 2005-12-20 2007-06-21 Roberts Douglas N Methods for assessment of native chromatin on microarrays
WO2007087310A2 (en) 2006-01-23 2007-08-02 Population Genetics Technologies Ltd. Nucleic acid analysis using sequence tokens
DK2385143T3 (en) 2006-02-02 2016-09-19 Univ Leland Stanford Junior Non-invasive fetal genetic screening by digital analysis
WO2007092538A2 (en) 2006-02-07 2007-08-16 President And Fellows Of Harvard College Methods for making nucleotide probes for sequencing and synthesis
GB0603251D0 (en) 2006-02-17 2006-03-29 Isis Innovation DNA conformation
WO2007111937A1 (en) 2006-03-23 2007-10-04 Applera Corporation Directed enrichment of genomic dna for high-throughput sequencing
RU2321638C2 (en) 2006-05-23 2008-04-10 Закрытое акционерное общество "Молекулярно-медицинские технологии" Method for preparing multifunctional multichip, multichip for successive or parallel screening biopolymers, method for analysis of biopolymers and set for realization of method
EP3424598B1 (en) 2006-06-14 2022-06-08 Verinata Health, Inc. Rare cell analysis using sample splitting and dna tags
US20080076909A1 (en) 2006-06-30 2008-03-27 Applera Corporation Emulsion pcr and amplicon capture
US8394590B2 (en) 2006-08-02 2013-03-12 California Institute Of Technology Capture agents and related methods and systems for detecting and/or sorting targets
JP5553602B2 (en) 2006-09-06 2014-07-16 キヤノン ユー.エス. ライフ サイエンシズ, インコーポレイテッド Chip and cartridge design configuration for performing microfluidic assays
WO2008037082A1 (en) 2006-09-25 2008-04-03 Archer-Daniels-Midland Company Superabsorbent surface-treated carboxyalkylated polysaccharides and process for producing same
US7935518B2 (en) 2006-09-27 2011-05-03 Alessandra Luchini Smart hydrogel particles for biomarker harvesting
US20080166720A1 (en) 2006-10-06 2008-07-10 The Regents Of The University Of California Method and apparatus for rapid nucleic acid analysis
WO2008061165A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic cartridge and method of making same
EP2109773A2 (en) * 2006-11-20 2009-10-21 Nativis, Inc Apparatus and method for transducing an in vitro or mammalian system with a low-frequency signal
US20080242560A1 (en) 2006-11-21 2008-10-02 Gunderson Kevin L Methods for generating amplified nucleic acid arrays
US8598328B2 (en) 2006-12-13 2013-12-03 National University Corporation Nagoya University Tol1 factor transposase and DNA introduction system using the same
EP2639579B1 (en) 2006-12-14 2016-11-16 Life Technologies Corporation Apparatus for measuring analytes using large scale FET arrays
US8338166B2 (en) * 2007-01-04 2012-12-25 Lawrence Livermore National Security, Llc Sorting, amplification, detection, and identification of nucleic acid subsequences in a complex mixture
JP2008167722A (en) 2007-01-15 2008-07-24 Konica Minolta Medical & Graphic Inc Nucleic acid isolation method by heating on magnetic support
US7844658B2 (en) 2007-01-22 2010-11-30 Comcast Cable Holdings, Llc System and method for providing an application to a device
EP2121983A2 (en) 2007-02-02 2009-11-25 Illumina Cambridge Limited Methods for indexing samples and sequencing multiple nucleotide templates
US20080228268A1 (en) 2007-03-15 2008-09-18 Uluru, Inc. Method of Formation of Viscous, Shape Conforming Gels and Their Uses as Medical Prosthesis
US7943330B2 (en) 2007-03-23 2011-05-17 Academia Sinica Tailored glycoproteomic methods for the sequencing, mapping and identification of cellular glycoproteins
US9222936B2 (en) 2007-04-18 2015-12-29 Solulink, Inc. Methods and/or use of oligonucleotide conjugates for suppressing background due to cross-hybridization
CN101293191B (en) 2007-04-25 2011-11-09 中国科学院过程工程研究所 Agarose gelatin microsphere preparation method
WO2008135512A2 (en) 2007-05-02 2008-11-13 Jerzy Paszkowski Dna amplification method
US20080295909A1 (en) 2007-05-24 2008-12-04 Locascio Laurie E Microfluidic Device for Passive Sorting and Storage of Liquid Plugs Using Capillary Force
US20090105959A1 (en) 2007-06-01 2009-04-23 Braverman Michael S System and method for identification of individual samples from a multiplex mixture
WO2008148200A1 (en) 2007-06-05 2008-12-11 Eugenia Kumacheva Multiple continuous microfluidic reactors for the scaled up synthesis of gel or polymer particles
US8454906B2 (en) 2007-07-24 2013-06-04 The Regents Of The University Of California Microfabricated droplet generator for single molecule/cell genetic analysis in engineered monodispersed emulsions
US20130084243A1 (en) 2010-01-27 2013-04-04 Liliane Goetsch Igf-1r specific antibodies useful in the detection and diagnosis of cellular proliferative disorders
WO2009023821A1 (en) 2007-08-15 2009-02-19 Opgen, Inc. Method, system and software arrangement for comparative analysis and phylogeny with whole-genome optical maps
WO2009048532A2 (en) 2007-10-05 2009-04-16 President And Fellows Of Harvard College Formation of particles for ultrasound application, drug release, and other uses, and microfluidic methods of preparation
US20090099040A1 (en) 2007-10-15 2009-04-16 Sigma Aldrich Company Degenerate oligonucleotides and their uses
US20100086914A1 (en) 2008-10-03 2010-04-08 Roche Molecular Systems, Inc. High resolution, high throughput hla genotyping by clonal sequencing
US8334013B2 (en) 2007-11-02 2012-12-18 Stc.Unm Mesoporous metal oxide microspheres and method for forming same
WO2009076485A2 (en) 2007-12-10 2009-06-18 Xiaolian Gao Sequencing of nucleic acids
US7771944B2 (en) 2007-12-14 2010-08-10 The Board Of Trustees Of The University Of Illinois Methods for determining genetic haplotypes and DNA mapping
KR20090081260A (en) 2008-01-23 2009-07-28 삼성전자주식회사 Assay method of microarray hybridization
AU2009212165A1 (en) 2008-02-07 2009-08-13 Pacific Biosciences Of California, Inc. CIS reactive oxygen quenchers integrated into linkers
US8034568B2 (en) 2008-02-12 2011-10-11 Nugen Technologies, Inc. Isothermal nucleic acid amplification methods and compositions
CN101918597B (en) 2008-03-11 2013-09-18 国立癌中心 Method for measuring chromosome, gene or specific nucleotide sequence copy numbers using SNP array
US9011777B2 (en) 2008-03-21 2015-04-21 Lawrence Livermore National Security, Llc Monodisperse microdroplet generation and stopping without coalescence
US8961902B2 (en) 2008-04-23 2015-02-24 Bioscale, Inc. Method and apparatus for analyte processing
DE102008025656B4 (en) 2008-05-28 2016-07-28 Genxpro Gmbh Method for the quantitative analysis of nucleic acids, markers therefor and their use
GB0810051D0 (en) 2008-06-02 2008-07-09 Oxford Biodynamics Ltd Method of diagnosis
US10722562B2 (en) 2008-07-23 2020-07-28 Immudex Aps Combinatorial analysis and repair
US20100062494A1 (en) 2008-08-08 2010-03-11 President And Fellows Of Harvard College Enzymatic oligonucleotide pre-adenylation
US9417190B2 (en) 2008-09-23 2016-08-16 Bio-Rad Laboratories, Inc. Calibrations and controls for droplet-based assays
US20120252015A1 (en) 2011-02-18 2012-10-04 Bio-Rad Laboratories Methods and compositions for detecting genetic material
US9764322B2 (en) 2008-09-23 2017-09-19 Bio-Rad Laboratories, Inc. System for generating droplets with pressure monitoring
US8663920B2 (en) 2011-07-29 2014-03-04 Bio-Rad Laboratories, Inc. Library characterization by digital assay
CA2735899A1 (en) 2008-09-25 2010-04-01 Cephalon, Inc. Liquid formulations of bendamustine
US8361299B2 (en) 2008-10-08 2013-01-29 Sage Science, Inc. Multichannel preparative electrophoresis system
ES2637843T3 (en) 2008-10-24 2017-10-17 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US9080211B2 (en) 2008-10-24 2015-07-14 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US20100113296A1 (en) 2008-11-05 2010-05-06 Joel Myerson Methods And Kits For Nucleic Acid Analysis
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US20100203647A1 (en) 2008-11-21 2010-08-12 The Rockefeller University Chemical Reporters of Protein Acylation
US20100136559A1 (en) 2008-12-02 2010-06-03 Bio-Rad Laboratories, Inc. Chromatin structure detection
US10839940B2 (en) 2008-12-24 2020-11-17 New York University Method, computer-accessible medium and systems for score-driven whole-genome shotgun sequence assemble
KR101065807B1 (en) 2009-01-23 2011-09-19 충남대학교산학협력단 Preparation method for micro-capsule using a microfluidic chip system
JP5457222B2 (en) * 2009-02-25 2014-04-02 エフ.ホフマン−ラ ロシュ アーゲー Miniaturized high-throughput nucleic acid analysis
US9347092B2 (en) 2009-02-25 2016-05-24 Roche Molecular System, Inc. Solid support for high-throughput nucleic acid analysis
WO2010104604A1 (en) 2009-03-13 2010-09-16 President And Fellows Of Harvard College Method for the controlled creation of emulsions, including multiple emulsions
EP2230312A1 (en) 2009-03-19 2010-09-22 Helmholtz-Zentrum für Infektionsforschung GmbH Probe compound for detecting and isolating enzymes and means and methods using the same
EP2411148B1 (en) 2009-03-23 2018-02-21 Raindance Technologies, Inc. Manipulation of microfluidic droplets
DK2414548T3 (en) 2009-03-30 2015-12-21 Illumina Inc Gene expression in single cells
SG10201402770YA (en) 2009-04-02 2014-08-28 Fluidigm Corp Multi-primer amplification method for barcoding of target nucleic acids
US9085798B2 (en) 2009-04-30 2015-07-21 Prognosys Biosciences, Inc. Nucleic acid constructs and methods of use
EP2427572B1 (en) 2009-05-01 2013-08-28 Illumina, Inc. Sequencing methods
US9334531B2 (en) 2010-12-17 2016-05-10 Life Technologies Corporation Nucleic acid amplification
FR2945545B1 (en) 2009-05-14 2011-08-05 Univ Aix Marseille Ii METHOD FOR DETECTION OF PROCARYOTE DNA EXTRACTED FROM A SAMPLE SAMPLE
FR2945819B1 (en) 2009-05-19 2011-06-17 Commissariat Energie Atomique DEVICE AND METHOD FOR ISOLATING BIOLOGICAL OR CHEMICAL TARGETS
US8574835B2 (en) 2009-05-29 2013-11-05 Life Technologies Corporation Scaffolded nucleic acid polymer particles and methods of making and using
US9524369B2 (en) 2009-06-15 2016-12-20 Complete Genomics, Inc. Processing and analysis of complex nucleic acid sequence data
US20110028412A1 (en) 2009-08-03 2011-02-03 Cappellos, Inc. Herbal enhanced analgesic formulations
US20110033548A1 (en) 2009-08-05 2011-02-10 E.I. Du Pont De Nemours And Company Degradable crosslinked aminated dextran microspheres and methods of use
EP3029141A1 (en) 2009-08-20 2016-06-08 Population Genetics Technologies Ltd. Compositions and methods for intramolecular nucleic acid rearrangement
JP2013503605A (en) 2009-09-01 2013-02-04 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Devices and methods for microarray selection
EP2940153B1 (en) 2009-09-02 2020-05-13 Bio-Rad Laboratories, Inc. System for mixing fluids by coalescence of multiple emulsions
US9625454B2 (en) 2009-09-04 2017-04-18 The Research Foundation For The State University Of New York Rapid and continuous analyte processing in droplet microfluidic devices
CA2816925C (en) 2009-11-04 2023-01-10 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
US9023769B2 (en) 2009-11-30 2015-05-05 Complete Genomics, Inc. cDNA library for nucleic acid sequencing
JP2013514079A (en) 2009-12-17 2013-04-25 キージーン・エン・フェー Whole genome sequencing based on restriction enzymes
CA2789425C (en) 2010-02-12 2020-04-28 Raindance Technologies, Inc. Digital analyte analysis with polymerase error correction
JP5901046B2 (en) 2010-02-19 2016-04-06 国立大学法人 千葉大学 Novel alternative splicing variant of OATP1B3 mRNA
US20110257889A1 (en) 2010-02-24 2011-10-20 Pacific Biosciences Of California, Inc. Sequence assembly and consensus sequence determination
EP2539450B1 (en) 2010-02-25 2016-02-17 Advanced Liquid Logic, Inc. Method of making nucleic acid libraries
US8236574B2 (en) 2010-03-01 2012-08-07 Quanterix Corporation Ultra-sensitive detection of molecules or particles using beads or other capture objects
WO2011119881A1 (en) 2010-03-24 2011-09-29 Northeastern University Multi-compartmental macrophage delivery
FR2958186A1 (en) 2010-03-30 2011-10-07 Ecole Polytech DEVICE FOR FORMING DROPS IN A MICROFLUID CIRCUIT.
WO2011140510A2 (en) 2010-05-06 2011-11-10 Bioo Scientific Corporation Oligonucleotide ligation, barcoding and methods and compositions for improving data quality and throughput using massively parallel sequencing
US20110287947A1 (en) 2010-05-18 2011-11-24 University Of Southern California Tethered Conformation Capture
JP6367554B2 (en) 2010-05-26 2018-08-01 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Dosage selection of adjuvanted synthetic nanocarriers
EP2580378A4 (en) 2010-06-08 2014-01-01 Nugen Technologies Inc Methods and composition for multiplex sequencing
US8703493B2 (en) 2010-06-15 2014-04-22 Src, Inc. Location analysis using fire retardant-protected nucleic acid-labeled tags
US20120003657A1 (en) 2010-07-02 2012-01-05 Samuel Myllykangas Targeted sequencing library preparation by genomic dna circularization
US20120034603A1 (en) 2010-08-06 2012-02-09 Tandem Diagnostics, Inc. Ligation-based detection of genetic variants
CN103202812B (en) 2010-08-09 2015-10-28 南京大学 A kind of method of protein nano grain for the preparation of sending pharmacological active substance in body
WO2012019765A1 (en) 2010-08-10 2012-02-16 European Molecular Biology Laboratory (Embl) Methods and systems for tracking samples and sample combinations
CN103328007B (en) 2010-09-16 2016-09-21 北卡罗来纳州大学查珀尔希尔分校 Supporting agent and the asymmetric difunctionality silyl group monomer of prodrug and granule thereof is delivered as pharmaceutical agent, chemical reagent and biological reagent
CN102409048B (en) * 2010-09-21 2013-10-23 深圳华大基因科技服务有限公司 DNA index library building method based on high throughput sequencing
US9187783B2 (en) 2010-10-04 2015-11-17 Genapsys, Inc. Systems and methods for automated reusable parallel biological reactions
EP2625295B1 (en) 2010-10-08 2019-03-13 President and Fellows of Harvard College High-throughput immune sequencing
CN102050953B (en) 2010-10-18 2012-11-07 武汉理工大学 Method for preparing reducible and degradable supermolecule hydrogel
US8753816B2 (en) 2010-10-26 2014-06-17 Illumina, Inc. Sequencing methods
US20130225623A1 (en) 2010-10-27 2013-08-29 Mount Sinai School Of Medicine Methods of Treating Psychiatric or Neurological Disorders with MGLUR Antagonists
CN103429331B (en) 2010-11-01 2016-09-28 伯乐生命医学产品有限公司 For forming the system of emulsion
US8829171B2 (en) 2011-02-10 2014-09-09 Illumina, Inc. Linking sequence reads using paired code tags
CA2821299C (en) 2010-11-05 2019-02-12 Frank J. Steemers Linking sequence reads using paired code tags
US9074251B2 (en) 2011-02-10 2015-07-07 Illumina, Inc. Linking sequence reads using paired code tags
CN102115789B (en) * 2010-12-15 2013-03-13 厦门大学 Nucleic acid label for second-generation high-flux sequencing and design method thereof
WO2012088348A2 (en) 2010-12-23 2012-06-28 Sequenom, Inc. Fetal genetic variation detection
US9163281B2 (en) 2010-12-23 2015-10-20 Good Start Genetics, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
WO2012100216A2 (en) 2011-01-20 2012-07-26 Knome, Inc. Methods and apparatus for assigning a meaningful numeric value to genomic variants, and searching and assessing same
GB201101429D0 (en) 2011-01-27 2011-03-16 Biocompatibles Uk Ltd Drug delivery system
DK3037536T3 (en) 2011-01-28 2020-01-13 Illumina Inc OLIGONUCLEOTID REPLACEMENT FOR DI-TAGGED AND DIRECTORY LIBRARIES
ES2687761T3 (en) 2011-01-31 2018-10-29 F. Hoffmann-La Roche Ag Methods of identification of multiple epitopes in cells
WO2012106546A2 (en) * 2011-02-02 2012-08-09 University Of Washington Through Its Center For Commercialization Massively parallel continguity mapping
EP3940084A1 (en) * 2011-02-09 2022-01-19 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
US9266104B2 (en) 2011-02-11 2016-02-23 Raindance Technologies, Inc. Thermocycling device for nucleic acid amplification and methods of use
WO2012109600A2 (en) 2011-02-11 2012-08-16 Raindance Technologies, Inc. Methods for forming mixed droplets
WO2012116250A1 (en) 2011-02-25 2012-08-30 University Of Massachusetts Medical School Monomers and polymers for functional polycarbonates and poly (ester-carbonates) and peg-co-polycarbonate hydrogels
WO2012116331A2 (en) 2011-02-25 2012-08-30 Illumina, Inc. Methods and systems for haplotype determination
CN103502218B (en) 2011-03-04 2016-08-17 生命科技公司 Compounds and methods for for conjugating biomolecules
US9215162B2 (en) 2011-03-09 2015-12-15 Annai Systems Inc. Biological data networks and methods therefor
JP2014509865A (en) * 2011-03-18 2014-04-24 バイオ−ラッド・ラボラトリーズ・インコーポレーテッド Multiplexed digital assay using a combination of signals
WO2012129363A2 (en) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
US20140141442A1 (en) 2011-04-05 2014-05-22 Institut National De La Sante Et De La Recherche Medicale (Inserm) Linear dna amplification
GB2489714B (en) 2011-04-05 2013-11-06 Tracesa Ltd Fluid Identification Method
JP2014516514A (en) 2011-04-14 2014-07-17 コンプリート・ジェノミックス・インコーポレイテッド Processing and analysis of complex nucleic acid sequence data
DK2702146T3 (en) 2011-04-28 2019-03-11 Univ Leland Stanford Junior IDENTIFICATION OF POLYNUCLEOTIDES CONNECTED WITH A SAMPLE
CN106912197B (en) 2011-04-28 2022-01-25 生命技术公司 Methods and compositions for multiplex PCR
WO2012156918A1 (en) 2011-05-16 2012-11-22 Koninklijke Philips Electronics N.V. Bio-orthogonal drug activation
US9005935B2 (en) 2011-05-23 2015-04-14 Agilent Technologies, Inc. Methods and compositions for DNA fragmentation and tagging by transposases
US9150916B2 (en) 2011-06-24 2015-10-06 Beat Christen Compositions and methods for identifying the essential genome of an organism
US8927218B2 (en) 2011-06-27 2015-01-06 Flir Systems, Inc. Methods and compositions for segregating target nucleic acid from mixed nucleic acid samples
US8975302B2 (en) 2011-07-07 2015-03-10 Life Technologies Corporation Polymer particles, nucleic acid polymer particles and methods of making and using the same
US20130017978A1 (en) 2011-07-11 2013-01-17 Finnzymes Oy Methods and transposon nucleic acids for generating a dna library
US9605304B2 (en) 2011-07-20 2017-03-28 The Hong Kong Polytechnic University Ultra-stable oligonucleotide-gold and-silver nanoparticle conjugates and method of their preparation
US8658430B2 (en) 2011-07-20 2014-02-25 Raindance Technologies, Inc. Manipulating droplet size
EP2739394A2 (en) 2011-08-04 2014-06-11 Sage Science, Inc. Microfluidic systems and methods for processing fluids
WO2013022961A1 (en) 2011-08-08 2013-02-14 3The Broad Institute Compositions and methods for co-amplifying subsequences of a nucleic acid fragment sequence
US8846883B2 (en) 2011-08-16 2014-09-30 University Of Southhampton Oligonucleotide ligation
WO2013035114A1 (en) 2011-09-08 2013-03-14 Decode Genetics Ehf Tp53 genetic variants predictive of cancer
LT2768607T (en) 2011-09-26 2021-12-27 Thermo Fisher Scientific Geneart Gmbh Multiwell plate for high efficiency, small volume nucleic acid synthesis
CN106423314B (en) 2011-09-28 2021-03-02 哈佛学院院长等 Systems and methods for droplet generation and/or fluid manipulation
WO2013055955A1 (en) 2011-10-12 2013-04-18 Complete Genomics, Inc. Identification of dna fragments and structural variations
US9469874B2 (en) 2011-10-18 2016-10-18 The Regents Of The University Of California Long-range barcode labeling-sequencing
US20130109576A1 (en) 2011-10-28 2013-05-02 Anthony P. Shuber Methods for detecting mutations
CN104012011B (en) 2011-11-04 2018-11-13 英特尔公司 The signaling of configuration for downlink multipoint cooperation communication
EP2786019B1 (en) 2011-11-16 2018-07-25 International Business Machines Corporation Microfluidic device with deformable valve
US10689643B2 (en) 2011-11-22 2020-06-23 Active Motif, Inc. Targeted transposition for use in epigenetic studies
US9938524B2 (en) 2011-11-22 2018-04-10 Active Motif, Inc. Multiplex isolation of protein-associated nucleic acids
WO2013082612A1 (en) 2011-12-03 2013-06-06 Emd Millipore Corporation Micro-incubation systems for microfluidic cell culture and methods
EP4108782B1 (en) 2011-12-22 2023-06-07 President and Fellows of Harvard College Compositions and methods for analyte detection
WO2013096643A1 (en) 2011-12-23 2013-06-27 Gigagen Methods and apparatuses for droplet mixing
KR102019297B1 (en) 2012-02-09 2019-09-06 라이프 테크놀로지스 코포레이션 Hydrophilic polymeric particles and methods for making same
EP2814984A4 (en) 2012-02-14 2015-07-29 Univ Johns Hopkins Mirna analysis methods
EP2814472A4 (en) 2012-02-15 2015-11-04 Wisconsin Alumni Res Found Dithioamine reducing agents
ES2855130T3 (en) 2012-02-17 2021-09-23 Hutchinson Fred Cancer Res Compositions and methods to accurately identify mutations
EP2823064B1 (en) * 2012-03-05 2019-02-06 President and Fellows of Harvard College Methods for epigenetic sequencing
NO2694769T3 (en) 2012-03-06 2018-03-03
US9552458B2 (en) 2012-03-16 2017-01-24 The Research Institute At Nationwide Children's Hospital Comprehensive analysis pipeline for discovery of human genetic variation
WO2013148189A1 (en) 2012-03-30 2013-10-03 Massachusetts Institute Of Technology Probe incorporation mediated by enzymes
EP2647426A1 (en) 2012-04-03 2013-10-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Replication of distributed nucleic acid molecules with preservation of their relative distribution through hybridization-based binding
US8209130B1 (en) 2012-04-04 2012-06-26 Good Start Genetics, Inc. Sequence assembly
BR112014025695B1 (en) 2012-04-16 2021-08-03 Biological Dynamics, Inc NUCLEIC ACID SAMPLE PREPARATION
US20130296173A1 (en) 2012-04-23 2013-11-07 Complete Genomics, Inc. Pre-anchor wash
CA2875695C (en) 2012-06-15 2022-11-15 The Board Of Regents Of The University Of Texas System High throughput sequencing of multiple transcripts of a single cell
WO2014018460A1 (en) 2012-07-24 2014-01-30 Sequenta, Inc. Single cell analysis using sequence tags
EP3578669A1 (en) 2012-08-08 2019-12-11 F. Hoffmann-La Roche AG Increasing dynamic range for identifying multiple epitopes in cells
CA2881783A1 (en) 2012-08-13 2014-02-20 The Regents Of The University Of California Methods and systems for detecting biological components
US10584381B2 (en) 2012-08-14 2020-03-10 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
WO2014047561A1 (en) * 2012-09-21 2014-03-27 The Broad Institute Inc. Compositions and methods for labeling of agents
US9644199B2 (en) * 2012-10-01 2017-05-09 Agilent Technologies, Inc. Immobilized transposase complexes for DNA fragmentation and tagging
GB201217772D0 (en) 2012-10-04 2012-11-14 Base4 Innovation Ltd Sequencing method
FR2996545B1 (en) 2012-10-08 2016-03-25 Ecole Polytech MICROFLUIDIC METHOD FOR PROCESSING AND ANALYZING A SOLUTION CONTAINING BIOLOGICAL MATERIAL, AND CORRESPONDING MICROFLUIDIC CIRCUIT
FR2996544B1 (en) 2012-10-08 2015-03-13 Ecole Polytech MICROFLUIDIC CIRCUIT FOR COMBINING DROPS OF MULTIPLE FLUIDS AND CORRESPONDING MICROFLUIDIC PROCESS.
EP2906715B1 (en) 2012-10-15 2017-07-26 Life Technologies Corporation Compositions, methods, systems and kits for target nucleic acid enrichment
CN107090491A (en) 2012-11-05 2017-08-25 鲁比康基因组学公司 Bar coding nucleic acid
EP2917366B1 (en) 2012-11-06 2017-08-02 Oxford Nanopore Technologies Limited Quadruplex method
WO2014074611A1 (en) 2012-11-07 2014-05-15 Good Start Genetics, Inc. Methods and systems for identifying contamination in samples
WO2014109845A1 (en) 2012-12-03 2014-07-17 Yilin Zhang Single-stranded polynucleotide amplification methods
EP2931899A1 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
EP4279588A3 (en) 2012-12-12 2024-01-17 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP2749653A1 (en) 2012-12-28 2014-07-02 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Molecular coding for analysis of composition of macromolecules and molecular complexes
EP2752664A1 (en) 2013-01-07 2014-07-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Label-free method for the detection of analytes
US9683230B2 (en) 2013-01-09 2017-06-20 Illumina Cambridge Limited Sample preparation on a solid support
EP2994749A4 (en) 2013-01-17 2017-07-19 Edico Genome Corp. Bioinformatics systems, apparatuses, and methods executed on an integrated circuit processing platform
KR101984699B1 (en) 2013-01-24 2019-05-31 삼성전자주식회사 Micro-fluidic system for analysis of nucleic acid
EP3473905B1 (en) 2013-01-25 2020-07-29 Bio-rad Laboratories, Inc. System and method for performing droplet inflation
US10381106B2 (en) 2013-01-28 2019-08-13 Hasso-Plattner-Institut Fuer Softwaresystemtechnik Gmbh Efficient genomic read alignment in an in-memory database
US11110458B2 (en) 2013-02-01 2021-09-07 Bio-Rad Laboratories, Inc. System for detection of spaced droplets
JP2016514047A (en) 2013-03-06 2016-05-19 プレジデント アンド フェローズ オブ ハーバード カレッジ Device and method for forming relatively monodisperse droplets
SG11201507087WA (en) 2013-03-08 2015-10-29 Bio Rad Laboratories Compositions, methods and systems for polymerase chain reaction assays
WO2014142850A1 (en) 2013-03-13 2014-09-18 Illumina, Inc. Methods and compositions for nucleic acid sequencing
WO2014152155A1 (en) 2013-03-14 2014-09-25 The Broad Institute, Inc. Massively multiplexed rna sequencing
US9273349B2 (en) 2013-03-14 2016-03-01 Affymetrix, Inc. Detection of nucleic acids
CA3156663A1 (en) 2013-03-15 2014-09-18 Verinata Health, Inc. Generating cell-free dna libraries directly from blood
WO2014145760A1 (en) 2013-03-15 2014-09-18 Bio-Rad Laboratories, Inc. Droplet generator with collection tube
EP2971080B1 (en) 2013-03-15 2018-01-03 Expedeon, S.L. Methods for amplification and sequencing using thermostable tthprimpol
US9328382B2 (en) 2013-03-15 2016-05-03 Complete Genomics, Inc. Multiple tagging of individual long DNA fragments
CA2907050C (en) 2013-03-15 2023-09-26 Prognosys Biosciences, Inc. Methods for detecting peptide/mhc/tcr binding
US20140274729A1 (en) 2013-03-15 2014-09-18 Nugen Technologies, Inc. Methods, compositions and kits for generation of stranded rna or dna libraries
WO2014144495A1 (en) 2013-03-15 2014-09-18 Abvitro, Inc. Single cell bar-coding for antibody discovery
EP2981349A4 (en) 2013-04-02 2016-11-16 Raindance Technologies Inc Systems and methods for handling microfluidic droplets
EP2994559B1 (en) 2013-05-09 2020-07-08 Bio-rad Laboratories, Inc. Magnetic immuno digital pcr assay
EP2999792B1 (en) 2013-05-23 2018-11-14 The Board of Trustees of The Leland Stanford Junior University Transposition into native chromatin for personal epigenomics
WO2014201273A1 (en) 2013-06-12 2014-12-18 The Broad Institute, Inc. High-throughput rna-seq
JP6600302B2 (en) 2013-06-12 2019-10-30 ザ ジェネラル ホスピタル コーポレイション Methods, kits, and systems for multiplex detection of target molecules and uses thereof
BR112015031608A2 (en) 2013-06-17 2017-08-22 Massachusetts Inst Technology APPLICATION AND USE OF CRISPR-CAS SYSTEMS, VECTORS AND COMPOSITIONS FOR LIVER TARGETING AND THERAPY
US20160208323A1 (en) 2013-06-21 2016-07-21 The Broad Institute, Inc. Methods for Shearing and Tagging DNA for Chromatin Immunoprecipitation and Sequencing
CN110592182B (en) 2013-06-27 2023-11-28 10X基因组学有限公司 Compositions and methods for sample processing
CA2919503C (en) 2013-07-12 2019-06-25 University Of South Alabama Minimal piggybac vectors for genome integration
CN103394410B (en) 2013-07-25 2016-04-20 博奥生物集团有限公司 A kind of intelligent magnetic frame of position-adjustable
GB2516684A (en) 2013-07-30 2015-02-04 Sphere Fluidics Ltd Microfluidic devices and systems
KR102536833B1 (en) 2013-08-28 2023-05-26 벡톤 디킨슨 앤드 컴퍼니 Massively parallel single cell analysis
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
CA2959978A1 (en) 2013-09-24 2015-04-02 The Regents Of The University Of California Encapsulated sensors and sensing systems for bioassays and diagnostics and methods for making and using them
GB201317301D0 (en) 2013-09-30 2013-11-13 Linnarsson Sten Method for capturing and encoding nucleic acid from a plurality of single cells
US10266904B2 (en) 2013-10-09 2019-04-23 Stc.Unm Synthetic long read DNA sequencing
US9937495B2 (en) 2013-10-28 2018-04-10 Massachusetts Institute Of Technology Hydrogel microstructures with immiscible fluid isolation for small reaction volumes
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US20140315755A1 (en) 2013-12-26 2014-10-23 Tao Chen Genome-wide Antisense Oligonucleotide and RNAi
CN114717291A (en) 2013-12-30 2022-07-08 阿特雷卡公司 Analysis of nucleic acids associated with single cells using nucleic acid barcodes
KR101464100B1 (en) 2014-01-29 2014-11-21 성균관대학교산학협력단 Fusion nano liposome-fluorescence labeled nucleic acid for in vivo application, uses thereof and preparation method thereof
WO2015113725A1 (en) 2014-02-03 2015-08-06 Thermo Fisher Scientific Baltics Uab Method for controlled dna fragmentation
CN105531408B (en) 2014-02-13 2019-09-10 生物辐射实验室股份有限公司 Chromosomal conformation divides product capture
EP3110975B1 (en) 2014-02-27 2021-07-07 Jumpcode Genomics, Inc. Methods for analysis of somatic mobile elements, and uses thereof
EP3845640A3 (en) 2014-04-15 2021-09-01 Illumina, Inc. Modified tranposases for improved insertion sequence bias and increased dna input tolerence
DK3456846T3 (en) 2014-04-21 2022-07-11 Harvard College SYSTEMS AND METHODS FOR BARCODE MARKING NUCLEIC ACID
US20150298091A1 (en) 2014-04-21 2015-10-22 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
US10975371B2 (en) 2014-04-29 2021-04-13 Illumina, Inc. Nucleic acid sequence analysis from single cells
EP3146046B1 (en) 2014-05-23 2020-03-11 Digenomix Corporation Haploidome determination by digitized transposons
EP3152232B1 (en) 2014-06-06 2019-11-13 Herlev Hospital Determining antigen recognition through barcoding of mhc multimers
AU2015273480A1 (en) 2014-06-11 2016-12-08 Samplix S.A.R.L. Nucleotide sequence exclusion enrichment by droplet sorting (needls)
WO2015191877A1 (en) 2014-06-11 2015-12-17 Life Technologies Corporation Systems and methods for substrate enrichment
WO2015188839A2 (en) 2014-06-13 2015-12-17 Immudex Aps General detection and isolation of specific cells by binding of labeled molecules
US10480021B2 (en) 2014-06-23 2019-11-19 Yale University Methods for closed chromatin mapping and DNA methylation analysis for single cells
US11155809B2 (en) 2014-06-24 2021-10-26 Bio-Rad Laboratories, Inc. Digital PCR barcoding
SG11201610691QA (en) 2014-06-26 2017-01-27 10X Genomics Inc Processes and systems for nucleic acid sequence assembly
MX2016016898A (en) 2014-06-26 2017-04-25 10X Genomics Inc Methods and compositions for sample analysis.
US10017759B2 (en) 2014-06-26 2018-07-10 Illumina, Inc. Library preparation of tagged nucleic acid
WO2015200893A2 (en) 2014-06-26 2015-12-30 10X Genomics, Inc. Methods of analyzing nucleic acids from individual cells or cell populations
CN106536756A (en) 2014-06-26 2017-03-22 10X基因组学有限公司 Analysis of nucleic acid sequences
DK3656875T3 (en) 2014-07-18 2021-12-13 Illumina Inc Non-invasive prenatal diagnosis
US20160024558A1 (en) 2014-07-23 2016-01-28 10X Genomics, Inc. Nucleic acid binding proteins and uses thereof
CN105335116B (en) 2014-07-30 2018-11-09 联想(北京)有限公司 A kind of display control method and electronic equipment
EP3186418A2 (en) 2014-08-26 2017-07-05 Nugen Technologies, Inc. Compositions and methods for targeted nucleic acid sequence enrichment and high efficiency library generation
EP3191605B1 (en) 2014-09-09 2022-07-27 The Broad Institute, Inc. A droplet-based method and apparatus for composite single-cell nucleic acid analysis
ES2727656T3 (en) 2014-09-15 2019-10-17 Abvitro Llc High performance sequencing of nucleotide banks
BR122021026779B1 (en) 2014-10-17 2023-12-19 Illumina Cambridge Limited CONTIGUITY PRESERVING TRANSPOSON
CN107002128A (en) 2014-10-29 2017-08-01 10X 基因组学有限公司 The method and composition being sequenced for target nucleic acid
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
JP2017537645A (en) 2014-12-19 2017-12-21 アプライズ バイオ, インコーポレイテッド Method for identifying multiple epitopes in a selected subpopulation of cells
SG11201705615UA (en) 2015-01-12 2017-08-30 10X Genomics Inc Processes and systems for preparing nucleic acid sequencing libraries and libraries prepared using same
CA2968417A1 (en) 2015-01-13 2016-07-21 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
CA2975852A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
US11111519B2 (en) 2015-02-04 2021-09-07 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
CN107208156B (en) 2015-02-09 2021-10-08 10X基因组学有限公司 System and method for determining structural variation and phasing using variation recognition data
KR20210135626A (en) 2015-02-10 2021-11-15 일루미나, 인코포레이티드 The method and the composition for analyzing the cellular constituent
CA2975958A1 (en) 2015-02-24 2016-09-01 10X Genomics, Inc. Methods for targeted nucleic acid sequence coverage
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
CN107208158B (en) 2015-02-27 2022-01-28 贝克顿迪金森公司 Spatially addressable molecular barcode
EP3268462B1 (en) 2015-03-11 2021-08-11 The Broad Institute, Inc. Genotype and phenotype coupling
WO2016149661A1 (en) 2015-03-18 2016-09-22 The Broad Institute, Inc. Massively parallel on-chip coalescence of microemulsions
WO2016160908A1 (en) 2015-03-30 2016-10-06 Verily Life Sciences Llc Methods for combining single cell profiling with combinatorial nanoparticle conjugate library screening
JP2018511341A (en) 2015-04-17 2018-04-26 プレジデント アンド フェローズ オブ ハーバード カレッジ Barcoding systems and methods for gene sequencing and other applications
CN107250447B (en) 2015-04-20 2020-05-05 深圳华大生命科学研究院 Long fragment DNA library construction method
EP3708256B1 (en) 2015-04-22 2022-11-30 Stilla Technologies Contact-less priming method for loading a solution in a microfluidic device and associated system
US20160314242A1 (en) 2015-04-23 2016-10-27 10X Genomics, Inc. Sample indexing methods and compositions for sequencing applications
US20160348093A1 (en) 2015-05-18 2016-12-01 10X Genomics, Inc. Mobile Solid Phase Compositions for Use in Biochemical Reactions and Analyses
EP3298168A4 (en) 2015-05-18 2019-02-20 10X Genomics, Inc. Stabilized reducing agents and methods using same
EP3304383B1 (en) 2015-05-26 2021-07-07 Pacific Biosciences of California, Inc. De novo diploid genome assembly and haplotype sequence reconstruction
WO2016187717A1 (en) 2015-05-26 2016-12-01 Exerkine Corporation Exosomes useful for genome editing
US20180087050A1 (en) 2015-05-27 2018-03-29 Jianbiao Zheng Methods of inserting molecular barcodes
EP3822367A1 (en) 2015-06-24 2021-05-19 Oxford BioDynamics PLC Detection processes using sites of chromosome interaction
US10894980B2 (en) 2015-07-17 2021-01-19 President And Fellows Of Harvard College Methods of amplifying nucleic acid sequences mediated by transposase/transposon DNA complexes
PL3334841T3 (en) 2015-08-12 2020-05-18 Cemm - Forschungszentrum Für Molekulare Medizin Gmbh Methods for studying nucleic acids
WO2017034970A1 (en) 2015-08-21 2017-03-02 The General Hospital Corporation Combinatorial single molecule analysis of chromatin
US11156611B2 (en) 2015-09-24 2021-10-26 Abvitro Llc Single cell characterization using affinity-oligonucleotide conjugates and vessel barcoded polynucleotides
EP3366818B1 (en) 2015-10-19 2021-04-14 Zhejiang Annoroad Bio-Technology Co., Ltd. Method for constructing high-resolution single cell hi-c library with a lot of information
WO2017070056A1 (en) 2015-10-20 2017-04-27 10X Genomics, Inc. Methods and systems for high throughput single cell genetic manipulation
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
WO2017075294A1 (en) 2015-10-28 2017-05-04 The Board Institute Inc. Assays for massively combinatorial perturbation profiling and cellular circuit reconstruction
EP3371309B1 (en) 2015-11-04 2023-07-05 Atreca, Inc. Combinatorial sets of nucleic acid barcodes for analysis of nucleic acids associated with single cells
SG11201803983UA (en) 2015-11-19 2018-06-28 10X Genomics Inc Transformable tagging compositions, methods, and processes incorporating same
EP4144861A1 (en) 2015-12-04 2023-03-08 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
EP3397764A4 (en) 2015-12-30 2019-05-22 Bio-Rad Laboratories, Inc. Digital protein quantification
CN108779491B (en) 2016-02-11 2021-03-09 10X基因组学有限公司 Systems, methods, and media for de novo assembly of whole genome sequence data
US20170260584A1 (en) 2016-02-11 2017-09-14 10X Genomics, Inc. Cell population analysis using single nucleotide polymorphisms from single cell transcriptomes
US20190078150A1 (en) 2016-03-01 2019-03-14 Universal Sequencing Technology Corporation Methods and Kits for Tracking Nucleic Acid Target Origin for Nucleic Acid Sequencing
EP3426774A4 (en) 2016-03-10 2019-08-14 The Board of Trustees of the Leland Stanford Junior University Transposase-mediated imaging of the accessible genome
EP3443123B1 (en) 2016-04-11 2021-08-11 Board of Regents, The University of Texas System Methods and compositions for detecting single t cell receptor affinity and sequence
WO2017197343A2 (en) 2016-05-12 2017-11-16 10X Genomics, Inc. Microfluidic on-chip filters
WO2017197338A1 (en) 2016-05-13 2017-11-16 10X Genomics, Inc. Microfluidic systems and methods of use
RU2019106038A (en) 2016-08-10 2020-09-17 Президент Энд Фэллоуз Оф Харвард Коллидж DE NOVO METHODS FOR ASSEMBLING BARCODED GENOMIC DNA FRAGMENTS
US10688494B2 (en) 2016-08-23 2020-06-23 10X Genomics, Inc. Microfluidic surface-mediated emulsion stability control
WO2018044831A1 (en) 2016-08-30 2018-03-08 Integrated Dna Technologies, Inc. Cleavable hairpin primers
GB2569252A (en) 2016-08-31 2019-06-12 Harvard College Methods of combining the detection of biomolecules into a single assay using fluorescent in situ sequencing
RU2736351C2 (en) 2016-08-31 2020-11-16 Президент Энд Фэллоуз Оф Харвард Коллидж Methods for discrete amplification of complete genome
US20180080021A1 (en) 2016-09-17 2018-03-22 The Board Of Trustees Of The Leland Stanford Junior University Simultaneous sequencing of rna and dna from the same sample
US11460468B2 (en) 2016-09-26 2022-10-04 Becton, Dickinson And Company Measurement of protein expression using reagents with barcoded oligonucleotide sequences
DK3529357T3 (en) 2016-10-19 2022-04-25 10X Genomics Inc Methods for bar coding nucleic acid molecules from individual cells
ES2922281T3 (en) 2016-12-07 2022-09-12 Mgi Tech Co Ltd Method for constructing a single cell sequencing library and use thereof
WO2018118971A1 (en) 2016-12-19 2018-06-28 Bio-Rad Laboratories, Inc. Droplet tagging contiguity preserved tagmented dna
US20220033893A1 (en) 2016-12-21 2022-02-03 The Regents Of The University Of California Single Cell Genomic Sequencing Using Hydrogel Based Droplets
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US20190177800A1 (en) 2017-12-08 2019-06-13 10X Genomics, Inc. Methods and compositions for labeling cells
EP3565904A1 (en) 2016-12-29 2019-11-13 Illumina, Inc. Analysis system for orthogonal access to and tagging of biomolecules in cellular compartments
EP4095263A1 (en) 2017-01-06 2022-11-30 Editas Medicine, Inc. Methods of assessing nuclease cleavage
US20210087610A9 (en) 2017-01-12 2021-03-25 Massachusetts Institute Of Technology Methods for analyzing t cell receptors and b cell receptors
WO2018140966A1 (en) 2017-01-30 2018-08-02 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10347365B2 (en) 2017-02-08 2019-07-09 10X Genomics, Inc. Systems and methods for visualizing a pattern in a dataset
GB201704402D0 (en) 2017-03-20 2017-05-03 Blacktrace Holdings Ltd Single cell DNA sequencing
KR20190133711A (en) 2017-03-24 2019-12-03 싱가포르국립대학교 Multiple Detection Method of Molecules
EP3610034B1 (en) 2017-04-12 2022-06-08 Karius, Inc. Sample preparation methods, systems and compositions
WO2018191701A1 (en) 2017-04-14 2018-10-18 The Broad Institute, Inc. High-throughput screens for exploring biological functions of microscale biological systems
US20180312822A1 (en) 2017-04-26 2018-11-01 10X Genomics, Inc. Mmlv reverse transcriptase variants
CN110945139B (en) 2017-05-18 2023-09-05 10X基因组学有限公司 Method and system for sorting droplets and beads
US10544413B2 (en) 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
CN110870018A (en) 2017-05-19 2020-03-06 10X基因组学有限公司 System and method for analyzing a data set
US10914729B2 (en) 2017-05-22 2021-02-09 The Trustees Of Princeton University Methods for detecting protein binding sequences and tagging nucleic acids
US10844372B2 (en) 2017-05-26 2020-11-24 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
WO2018226546A1 (en) 2017-06-05 2018-12-13 10X Genomics, Inc. Gaskets for the distribution of pressures in a microfluidic system
US10676779B2 (en) 2017-06-05 2020-06-09 Becton, Dickinson And Company Sample indexing for single cells
SG11201912824YA (en) 2017-06-20 2020-01-30 10X Genomics Inc Methods and systems for improved droplet stabilization
EP3642358A1 (en) 2017-06-21 2020-04-29 Bluedot LLC Systems and methods for identification of nucleic acids in a sample
CN108336542B (en) 2017-06-23 2020-02-21 番禺得意精密电子工业有限公司 Electrical connector
NZ759924A (en) 2017-08-01 2023-07-28 Illumina Inc Hydrogel beads for nucleotide sequencing
US9946577B1 (en) 2017-08-14 2018-04-17 10X Genomics, Inc. Systems and methods for distributed resource management
US10821442B2 (en) 2017-08-22 2020-11-03 10X Genomics, Inc. Devices, systems, and kits for forming droplets
US10590244B2 (en) 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
EP3691703A1 (en) 2017-10-04 2020-08-12 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
WO2019084043A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods and systems for nuclecic acid preparation and chromatin analysis
WO2019083852A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Microfluidic channel networks for partitioning
US20190127731A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods for preparing nucleic acid molecules
WO2019084328A1 (en) 2017-10-26 2019-05-02 10X Genomics, Inc. Methods for preparing nucleic acid molecules
EP3700672B1 (en) 2017-10-27 2022-12-28 10X Genomics, Inc. Methods for sample preparation and analysis
SG11201913654QA (en) 2017-11-15 2020-01-30 10X Genomics Inc Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
WO2019108851A1 (en) 2017-11-30 2019-06-06 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
WO2019113235A1 (en) 2017-12-06 2019-06-13 10X Genomics, Inc. Methods and systems for processing nucleic acid molecules
EP3724658A1 (en) 2017-12-12 2020-10-21 10X Genomics, Inc. Systems and methods for single cell processing
CN111712579A (en) 2017-12-22 2020-09-25 10X基因组学有限公司 Systems and methods for processing nucleic acid molecules from one or more cells
WO2019148042A1 (en) 2018-01-26 2019-08-01 10X Genomics, Inc. Compositions and methods for sample processing
CN112004920A (en) 2018-02-05 2020-11-27 斯坦福大学托管董事会 System and method for multiple measurements of single cells and aggregated cells
EP3752832A1 (en) 2018-02-12 2020-12-23 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
EP3755812A1 (en) 2018-02-22 2020-12-30 10X Genomics, Inc. Ligation mediated analysis of nucleic acids
WO2019169028A1 (en) 2018-02-28 2019-09-06 10X Genomics, Inc. Transcriptome sequencing through random ligation
WO2019169347A1 (en) 2018-03-02 2019-09-06 10X Genomics, Inc. Systems and apparatus for holding plates
WO2019191321A1 (en) 2018-03-28 2019-10-03 10X Genomics, Inc. Nucleic acid enrichment within partitions
CN112262218A (en) 2018-04-06 2021-01-22 10X基因组学有限公司 System and method for quality control in single cell processing
WO2019217758A1 (en) 2018-05-10 2019-11-14 10X Genomics, Inc. Methods and systems for molecular library generation
US20190345636A1 (en) 2018-05-10 2019-11-14 10X Genomics, Inc. Methods and systems for molecular library generation
US20190352717A1 (en) 2018-05-18 2019-11-21 10X Genomics, Inc. Targeted non-invasive prenatal testing
US11932899B2 (en) 2018-06-07 2024-03-19 10X Genomics, Inc. Methods and systems for characterizing nucleic acid molecules
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
WO2020006183A1 (en) 2018-06-28 2020-01-02 10X Genomics, Inc. Systems and methods for visualization of single-cell resolution characteristics
US20200033366A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
US20200032335A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
WO2020028882A1 (en) 2018-08-03 2020-02-06 10X Genomics, Inc. Methods and systems to minimize barcode exchange
US20200056223A1 (en) 2018-08-20 2020-02-20 10X Genomics, Inc. Compositions and methods for cellular processing
EP3906318A1 (en) 2019-01-06 2021-11-10 10X Genomics, Inc. Methods and systems for enrichment of barcodes

Patent Citations (321)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0249007A2 (en) 1986-04-14 1987-12-16 The General Hospital Corporation A method of screening hybridomas
US5618711A (en) 1986-08-22 1997-04-08 Hoffmann-La Roche Inc. Recombinant expression vectors and purification methods for Thermus thermophilus DNA polymerase
US5202231A (en) 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
US5695940A (en) 1987-04-01 1997-12-09 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US5149625A (en) 1987-08-11 1992-09-22 President And Fellows Of Harvard College Multiplex analysis of DNA
US5994056A (en) 1991-05-02 1999-11-30 Roche Molecular Systems, Inc. Homogeneous methods for nucleic acid amplification and detection
US5413924A (en) 1992-02-13 1995-05-09 Kosak; Kenneth M. Preparation of wax beads containing a reagent for release by heating
US5436130A (en) 1992-03-19 1995-07-25 The Regents Of The University Of California Multiple tag labeling method for DNA sequencing
US5587128A (en) 1992-05-01 1996-12-24 The Trustees Of The University Of Pennsylvania Mesoscale polynucleotide amplification devices
EP0637996B1 (en) 1992-05-01 1997-07-23 The Trustees Of The University Of Pennsylvania Microfabricated detection structures
US5512131A (en) 1993-10-04 1996-04-30 President And Fellows Of Harvard College Formation of microstamped patterns on surfaces and derivative articles
US20030044777A1 (en) 1993-10-28 2003-03-06 Kenneth L. Beattie Flowthrough devices for multiple discrete binding reactions
US5834197A (en) 1994-05-11 1998-11-10 Genera Technologies Limited Methods of capturing species from liquids and assay procedures
US6172218B1 (en) 1994-10-13 2001-01-09 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
WO1996029629A2 (en) 1995-03-01 1996-09-26 President And Fellows Of Harvard College Microcontact printing on surfaces and derivative articles
EP1967592B1 (en) 1995-06-07 2010-04-28 Solexa, Inc. Method of improving the efficiency of polynucleotide sequencing
WO1996041011A1 (en) 1995-06-07 1996-12-19 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
US5851769A (en) 1995-09-27 1998-12-22 The Regents Of The University Of California Quantitative DNA fiber mapping
US6057107A (en) 1995-10-11 2000-05-02 Luminex Corporation Methods and compositions for flow cytometric determination of DNA sequences
US5736330A (en) 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US6051377A (en) 1995-11-30 2000-04-18 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
US6046003A (en) 1995-11-30 2000-04-04 Pharmaseq, Inc. Method of determining the sequence of nucleic acids employing solid-phase particles carrying transponders
US6361950B1 (en) 1995-11-30 2002-03-26 Pharmaseq, Inc. Multiplex assay for nucleic acids employing transponders
US20010044109A1 (en) 1995-11-30 2001-11-22 Pharmaseq, Inc. Method of determining the sequence of nucleic acids employing solid-phase particles carrying transponders
US6355198B1 (en) 1996-03-15 2002-03-12 President And Fellows Of Harvard College Method of forming articles including waveguides via capillary micromolding and microtransfer molding
US5958703A (en) 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
US20050042625A1 (en) 1997-01-15 2005-02-24 Xzillion Gmbh & Co. Mass label linked hybridisation probes
US20030108897A1 (en) 1997-01-16 2003-06-12 Drmanac Radoje T. Methods and compositions for detection or quantification of nucleic acid species
US6297006B1 (en) 1997-01-16 2001-10-02 Hyseq, Inc. Methods for sequencing repetitive sequences and for determining the order of sequence subfragments
US20020034737A1 (en) 1997-03-04 2002-03-21 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US6327410B1 (en) 1997-03-14 2001-12-04 The Trustees Of Tufts College Target analyte sensors utilizing Microspheres
US20050130188A1 (en) 1997-03-14 2005-06-16 The Trustees Of Tufts College Methods for detecting target analytes and enzymatic reactions
US6859570B2 (en) 1997-03-14 2005-02-22 Trustees Of Tufts College, Tufts University Target analyte sensors utilizing microspheres
US6670133B2 (en) 1997-04-04 2003-12-30 Caliper Technologies Corp. Microfluidic device for sequencing by hybridization
US20030104466A1 (en) 1997-04-04 2003-06-05 Caliper Technologies Corporation Microfluidic sequencing systems
US8067159B2 (en) 1997-04-17 2011-11-29 Applied Biosystems, Llc Methods of detecting amplified product
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
US7972778B2 (en) 1997-04-17 2011-07-05 Applied Biosystems, Llc Method for detecting the presence of a single target nucleic acid in a sample
US8278071B2 (en) 1997-04-17 2012-10-02 Applied Biosystems, Llc Method for detecting the presence of a single target nucleic acid in a sample
US20080213766A1 (en) 1997-04-17 2008-09-04 Cytonix Method and device for detecting the presence of a single target nucleic acid in samples
US6406848B1 (en) 1997-05-23 2002-06-18 Lynx Therapeutics, Inc. Planar arrays of microparticle-bound polynucleotides
US20020051992A1 (en) 1997-05-23 2002-05-02 Lynx Therapeutics, Inc. System and apparatus for sequential processing of analytes
US7282370B2 (en) 1997-05-23 2007-10-16 Solexa, Inc. System and apparatus for sequential processing of analytes
US6806052B2 (en) 1997-05-23 2004-10-19 Lynx Therapeutics, Inc. Planar arrays of microparticle-bound polynucleotides
US20090143244A1 (en) 1997-05-23 2009-06-04 Solexa, Inc. System and apparatus for sequential processing of analytes
EP2258846A2 (en) 1997-07-07 2010-12-08 Medical Research Council A method for increasing the concentration of a nucleic acid molecule
EP1908832B1 (en) 1997-07-07 2012-12-26 Medical Research Council A method for increasing the concentration of a nucleic acid molecule
EP1019496B1 (en) 1997-07-07 2004-09-29 Medical Research Council In vitro sorting method
EP1482036B1 (en) 1997-07-07 2007-10-03 Medical Research Council A method for increasing the concentration of a nucleic acid molecule
US7638276B2 (en) 1997-07-07 2009-12-29 454 Life Sciences Corporation In vitro sorting method
US6297017B1 (en) 1997-07-11 2001-10-02 Brax Group Limited Categorising nucleic acids
WO1999009217A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
US20010020588A1 (en) 1997-09-15 2001-09-13 Whitehead Institute For Biomedical Research Methods and apparatus for processing a sample of biomolecular analyte using a microfabricated device
US20020092767A1 (en) 1997-09-19 2002-07-18 Aclara Biosciences, Inc. Multiple array microfluidic device units
US6103537A (en) 1997-10-02 2000-08-15 Aclara Biosciences, Inc. Capillary assays involving separation of free and bound species
US6485944B1 (en) 1997-10-10 2002-11-26 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6511803B1 (en) 1997-10-10 2003-01-28 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6432360B1 (en) 1997-10-10 2002-08-13 President And Fellows Of Harvard College Replica amplification of nucleic acid arrays
US6929859B2 (en) 1997-10-14 2005-08-16 Don J. Chandler Precision fluorescently dyed particles and methods of making and using same
US20030028981A1 (en) 1997-10-14 2003-02-13 Chandler Don J. Precision fluorescently dyed particles and methods of making and using same
US6913935B1 (en) 1997-12-04 2005-07-05 Amersham Biosciences Uk Limited Multiple assay method
WO1999052708A1 (en) 1998-04-13 1999-10-21 Luminex Corporation Liquid labeling with fluorescent microparticles
US7645596B2 (en) 1998-05-01 2010-01-12 Arizona Board Of Regents Method of determining the nucleotide sequence of oligonucleotides and DNA molecules
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US20070020617A1 (en) 1998-08-07 2007-01-25 Cellay, Llc C/O One Cell Systems, Inc. Gel microdrops in genetic analysis
US20030207260A1 (en) 1998-08-07 2003-11-06 Cellay, Llc C/O One Cell Systems, Inc. Gel microdroplets in genetic analysis
WO2000008212A1 (en) 1998-08-07 2000-02-17 Cellay, Llc Gel microdrops in genetic analysis
US6586176B1 (en) 1998-08-07 2003-07-01 Cellay, Llc Gel microdrops in genetic analysis
US20030044836A1 (en) 1998-10-15 2003-03-06 Princeton University, Office Of Technology & Trademark Licensing Quantitative analysis of hybridization patterns and intensities in oligonucleotide arrays
WO2000026412A1 (en) 1998-11-02 2000-05-11 Kenneth Loren Beattie Nucleic acid analysis using sequence-targeted tandem hybridization
EP1905828B1 (en) 1999-01-07 2012-08-08 Medical Research Council Optical sorting method
US20040063138A1 (en) 1999-02-16 2004-04-01 Mcginnis Malcolm D. Polynucleotide sequencing method
US20030215862A1 (en) 1999-02-23 2003-11-20 Caliper Technologies Corp. Sequencing by incorporation
US6632655B1 (en) 1999-02-23 2003-10-14 Caliper Technologies Corp. Manipulation of microparticles in microfluidic systems
US20030008323A1 (en) 1999-04-15 2003-01-09 Ilya Ravkin Chemical-library composition and method
US20020179849A1 (en) 1999-05-12 2002-12-05 Kevin Maher Multiplexed fluorescent detection in microfluidic devices
US6372813B1 (en) 1999-06-25 2002-04-16 Motorola Methods and compositions for attachment of biomolecules to solid supports, hydrogels, and hydrogel arrays
US6524456B1 (en) 1999-08-12 2003-02-25 Ut-Battelle, Llc Microfluidic devices for the controlled manipulation of small volumes
WO2001014589A2 (en) 1999-08-20 2001-03-01 Luminex Corporation Liquid array technology
US20060292583A1 (en) 1999-08-30 2006-12-28 The Government of the U.S.A as represented by the Secretary of Dept. of Health and Human Services High speed parallel molecular nucleic acid sequencing
US7745178B2 (en) 1999-10-27 2010-06-29 Affymetrix, Inc. Complexity management of genomic DNA
US6974669B2 (en) 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US6800298B1 (en) 2000-05-11 2004-10-05 Clemson University Biological lubricant composition and method of applying lubricant composition
WO2001089787A2 (en) 2000-05-25 2001-11-29 President And Fellows Of Harvard College Microfluidic systems including three-dimensionally arrayed channel networks
US20060263888A1 (en) 2000-06-02 2006-11-23 Honeywell International Inc. Differential white blood count on a disposable card
US6632606B1 (en) 2000-06-12 2003-10-14 Aclara Biosciences, Inc. Methods for single nucleotide polymorphism detection
US20050244850A1 (en) 2000-06-21 2005-11-03 Hiu Huang Assembly of arrays on chips segmented from wafers
US20040132122A1 (en) 2000-06-21 2004-07-08 Sukanta Banerjee Multianalyte molecular analysis using application-specific random particle arrays
US8252539B2 (en) 2000-09-15 2012-08-28 California Institute Of Technology Microfabricated crossflow devices and methods
WO2002031203A2 (en) 2000-10-10 2002-04-18 Diversa Corporation High throughput or capillary-based screening for a bioactivity or biomolecule
US20020089100A1 (en) 2000-11-14 2002-07-11 Akira Kawasaki Production apparatus of monodisperse particle and production process of monodisperse particle and monodisperse particle produced by the process
US7268167B2 (en) 2001-02-23 2007-09-11 Japan Science And Technology Agency Process for producing emulsion and microcapsules and apparatus therefor
WO2002086148A1 (en) 2001-04-18 2002-10-31 Ambrigen, Llc Particle based assay system
US6806058B2 (en) 2001-05-26 2004-10-19 One Cell Systems, Inc. Secretions of proteins by encapsulated cells
US20050019839A1 (en) 2001-05-26 2005-01-27 Once Cell Systems, Inc. Secretions of proteins by encapsulated cells
US20030008285A1 (en) 2001-06-29 2003-01-09 Fischer Steven M. Method of DNA sequencing using cleavable tags
US6767731B2 (en) 2001-08-27 2004-07-27 Intel Corporation Electron induced fluorescent method for nucleic acid sequencing
US20030039978A1 (en) 2001-08-27 2003-02-27 Hannah Eric C. Electron induced fluorescent method for nucleic acid sequencing
US20030182068A1 (en) 2001-10-30 2003-09-25 Battersby Bronwyn J. Device and methods for directed synthesis of chemical libraries
US7622280B2 (en) 2001-11-16 2009-11-24 454 Life Sciences Corporation Emulsion compositions
US20030170698A1 (en) 2002-01-04 2003-09-11 Peter Gascoyne Droplet-based microfluidic oligonucleotide synthesis engine
US8889083B2 (en) 2002-05-09 2014-11-18 The University Of Chicago Device and method for pressure-driven plug transport and reaction
US7129091B2 (en) 2002-05-09 2006-10-31 University Of Chicago Device and method for pressure-driven plug transport and reaction
US8273573B2 (en) 2002-05-09 2012-09-25 The University Of Chicago Method for obtaining a collection of plugs comprising biological molecules
US8304193B2 (en) 2002-05-09 2012-11-06 The University Of Chicago Method for conducting an autocatalytic reaction in plugs in a microfluidic system
US8822148B2 (en) 2002-05-09 2014-09-02 The University Of Chicago Method of performing PCR reaction in continuously flowing microfluidic plugs
US8329407B2 (en) 2002-05-09 2012-12-11 The University Of Chicago Method for conducting reactions involving biological molecules in plugs in a microfluidic system
US8337778B2 (en) 2002-06-28 2012-12-25 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
US20050172476A1 (en) 2002-06-28 2005-08-11 President And Fellows Of Havard College Method and apparatus for fluid dispersion
JP2006507921A (en) 2002-06-28 2006-03-09 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Method and apparatus for fluid dispersion
WO2004002627A2 (en) 2002-06-28 2004-01-08 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
US7708949B2 (en) 2002-06-28 2010-05-04 President And Fellows Of Harvard College Method and apparatus for fluid dispersion
US20140037514A1 (en) 2002-06-28 2014-02-06 Governing Council of the Univ. of Toronto, the MaRS Centre Method and apparatus for fluid dispersion
WO2004010106A2 (en) 2002-07-24 2004-01-29 Ptc Therapeutics, Inc. METHODS FOR IDENTIFYING SMALL MOLEDULES THAT MODULATE PREMATURE TRANSLATION TERMINATION AND NONSENSE MEDIATED mRNA DECAY
US20060240506A1 (en) 2002-09-09 2006-10-26 Ariel Kushmaro Method for isolating and culturing unculturable microorganisms
US20090048124A1 (en) 2003-01-29 2009-02-19 Leamon John H Methods of amplifying and sequencing nucleic acids
US7842457B2 (en) 2003-01-29 2010-11-30 454 Life Sciences Corporation Bead emulsion nucleic acid amplification
US8765380B2 (en) 2003-01-29 2014-07-01 454 Life Sciences Corporation Bead emulsion nucleic acid amplification
US8748102B2 (en) 2003-01-29 2014-06-10 454 Life Sciences Corporation Bead emulsion nucleic acid amplification
EP2145955B1 (en) 2003-01-29 2012-02-22 454 Life Sciences Corporation Bead emulsion nucleic acid amplification
EP1594980B1 (en) 2003-01-29 2009-11-11 454 Corporation Bead emulsion nucleic acid amplification
US7323305B2 (en) 2003-01-29 2008-01-29 454 Life Sciences Corporation Methods of amplifying and sequencing nucleic acids
US20130078638A1 (en) 2003-01-29 2013-03-28 Jan Berka Bead Emulsion Nucleic Acid Amplification
US20110201526A1 (en) 2003-01-29 2011-08-18 Jan Berka Bead emulsion nucleic acid amplification
US20050079510A1 (en) 2003-01-29 2005-04-14 Jan Berka Bead emulsion nucleic acid amplification
US7041481B2 (en) 2003-03-14 2006-05-09 The Regents Of The University Of California Chemical amplification based on fluid partitioning
USRE41780E1 (en) 2003-03-14 2010-09-28 Lawrence Livermore National Security, Llc Chemical amplification based on fluid partitioning in an immiscible liquid
US20060153924A1 (en) 2003-03-31 2006-07-13 Medical Research Council Selection by compartmentalised screening
US20120010098A1 (en) 2003-03-31 2012-01-12 Medical Research Council Selection by compartmentalised screening
EP2540389A1 (en) 2003-03-31 2013-01-02 Medical Research Council Method of encapsulating a molecule and a microbead
US20100210479A1 (en) 2003-03-31 2010-08-19 Medical Research Council Method of synthesis and testing of cominatorial libraries using microcapsules
US20120010107A1 (en) 2003-03-31 2012-01-12 Medical Research Council Selection by compartmentalised screening
US20040258701A1 (en) 2003-04-04 2004-12-23 Pfizer Inc. Microfluidized oil-in-water emulsions and vaccine compositions
US20060163385A1 (en) 2003-04-10 2006-07-27 Link Darren R Formation and control of fluidic species
WO2004091763A2 (en) 2003-04-10 2004-10-28 President And Fellows Of Harvard College Formation and control of fluidic species
WO2004102204A1 (en) 2003-05-16 2004-11-25 Global Technologies (Nz) Ltd Method and apparatus for mixing sample and reagent in a suspension fluid
WO2004103565A2 (en) 2003-05-19 2004-12-02 Hans-Knöll-Institut für Naturstoff-Forschung e.V. Device and method for structuring liquids and for dosing reaction liquids into liquid compartments immersed in a separation medium
WO2004105734A1 (en) 2003-05-28 2004-12-09 Valorisation Recherche, Societe En Commandite Method of preparing microcapsules
US20090286687A1 (en) 2003-07-05 2009-11-19 The Johns Hopkins University Method and Compositions for Detection and Enumeration of Genetic Variations
WO2005021151A1 (en) 2003-08-27 2005-03-10 President And Fellows Of Harvard College Electronic control of fluidic species
US20070003442A1 (en) 2003-08-27 2007-01-04 President And Fellows Of Harvard College Electronic control of fluidic species
WO2005023331A2 (en) 2003-09-04 2005-03-17 The United States Of America As Represented By The Department Of Veterans Affairs Hydrogel nanocompsites for ophthalmic applications
US20110195496A1 (en) 2003-09-25 2011-08-11 Atsushi Muraguchi Microwell array chip and method of manufacturing same
WO2005040406A1 (en) 2003-10-17 2005-05-06 Diversa Corporation High throughput screening of antibody libraries
US20070077572A1 (en) 2003-11-24 2007-04-05 Yeda Research And Development Co. Ltd. Compositions and methods for in vitro sorting of molecular and cellular libraries
WO2005049787A2 (en) 2003-11-24 2005-06-02 Yeda Research And Development Co.Ltd. Compositions and methods for in vitro sorting of molecular and cellular libraries
US20050181379A1 (en) 2004-02-18 2005-08-18 Intel Corporation Method and device for isolating and positioning single nucleic acid molecules
WO2005082098A2 (en) 2004-02-27 2005-09-09 President And Fellows Of Harvard College Polony fluorescent in situ sequencing beads
US7425431B2 (en) 2004-02-27 2008-09-16 President And Fellows Of Harvard College Polony fluorescent in situ sequencing beads
US20050221339A1 (en) 2004-03-31 2005-10-06 Medical Research Council Harvard University Compartmentalised screening by microfluidic control
US20070092914A1 (en) 2004-03-31 2007-04-26 Medical Research Council, Harvard University Compartmentalised screening by microfluidic control
US20090197772A1 (en) 2004-03-31 2009-08-06 Andrew Griffiths Compartmentalised combinatorial chemistry by microfluidic control
US20060020371A1 (en) 2004-04-13 2006-01-26 President And Fellows Of Harvard College Methods and apparatus for manipulation and/or detection of biological samples and other objects
US7799553B2 (en) 2004-06-01 2010-09-21 The Regents Of The University Of California Microfabricated integrated DNA analysis system
US20050287572A1 (en) 2004-06-01 2005-12-29 The Regents Of The University Of California Microfabricated integrated DNA analysis system
US20070207060A1 (en) 2004-07-21 2007-09-06 Chengdu Kuachang Medical Industrial Limited Testing method of analytic chip of multiple reactors, the analytic chip, and the testing device
WO2006030993A1 (en) 2004-09-14 2006-03-23 Jin-Ho Choy Information code system using dna sequences
US20060073487A1 (en) 2004-10-01 2006-04-06 Oliver Kerry G System and method for inhibiting the decryption of a nucleic acid probe sequence used for the detection of a specific nucleic acid
US7968287B2 (en) 2004-10-08 2011-06-28 Medical Research Council Harvard University In vitro evolution in microfluidic systems
US9029083B2 (en) 2004-10-08 2015-05-12 Medical Research Council Vitro evolution in microfluidic systems
US20090197248A1 (en) 2004-10-08 2009-08-06 President And Fellows Of Harvard College Vitro evolution in microfluidic systems
US20130178368A1 (en) 2004-10-08 2013-07-11 Andrew David Griffiths In vitro evolution in microfluidic systems
US8871444B2 (en) 2004-10-08 2014-10-28 Medical Research Council In vitro evolution in microfluidic systems
US20060078888A1 (en) 2004-10-08 2006-04-13 Medical Research Council Harvard University In vitro evolution in microfluidic systems
WO2007001448A2 (en) 2004-11-04 2007-01-04 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US20080004436A1 (en) 2004-11-15 2008-01-03 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Directed Evolution and Selection Using in Vitro Compartmentalization
WO2006078841A1 (en) 2005-01-21 2006-07-27 President And Fellows Of Harvard College Systems and methods for forming fluidic droplets encapsulated in particles such as colloidal particles
US7604938B2 (en) 2005-02-18 2009-10-20 Canon U.S. Life Sciences, Inc. Devices and methods for monitoring genomic DNA of organisms
US20060257893A1 (en) 2005-02-18 2006-11-16 Toru Takahashi Devices and methods for monitoring genomic DNA of organisms
US20090025277A1 (en) 2005-02-21 2009-01-29 Kagoshima University Method for purifying biodiesel fuel
US20060199193A1 (en) 2005-03-04 2006-09-07 Tae-Woong Koo Sensor arrays and nucleic acid sequencing applications
WO2006096571A2 (en) 2005-03-04 2006-09-14 President And Fellows Of Harvard College Method and apparatus for forming multiple emulsions
US20070054119A1 (en) 2005-03-04 2007-03-08 Piotr Garstecki Systems and methods of forming particles
JP2006289250A (en) 2005-04-08 2006-10-26 Kao Corp Micro mixer and fluid mixing method using the same
US20090137414A1 (en) 2005-06-15 2009-05-28 Complete Genomics, Inc. Single molecule arrays for genetic and chemical analysis
US20110071053A1 (en) 2005-06-15 2011-03-24 Callida Genomics, Inc. Single Molecule Arrays for Genetic and Chemical Analysis
US7709197B2 (en) 2005-06-15 2010-05-04 Callida Genomics, Inc. Nucleic acid analysis by random mixtures of non-overlapping fragments
US7901891B2 (en) 2005-06-15 2011-03-08 Callida Genomics, Inc. Nucleic acid analysis by random mixtures of non-overlapping fragments
US20110319281A1 (en) 2005-06-15 2011-12-29 Callida Genomics, Inc. Nucleic Acid Analysis by Random Mixtures of Non-Overlapping Fragments
US20090011943A1 (en) 2005-06-15 2009-01-08 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20090137404A1 (en) 2005-06-15 2009-05-28 Complete Genomics, Inc. Single molecule arrays for genetic and chemical analysis
US20070099208A1 (en) 2005-06-15 2007-05-03 Radoje Drmanac Single molecule arrays for genetic and chemical analysis
US8133719B2 (en) 2005-06-15 2012-03-13 Callida Genomics, Inc. Methods for making single molecule arrays
US7536928B2 (en) 2005-06-16 2009-05-26 Ntn Corporation Ball screw
WO2007002490A2 (en) 2005-06-22 2007-01-04 The Research Foundation Of State University Of New York Massively parallel 2-dimensional capillary electrophoresis
US20070154903A1 (en) 2005-06-23 2007-07-05 Nanosphere, Inc. Selective isolation and concentration of nucleic acids from complex samples
US20100021973A1 (en) * 2005-08-02 2010-01-28 Makarov Vladimir L Compositions and methods for processing and amplification of dna, including using multiple enzymes in a single reaction
WO2007024840A2 (en) 2005-08-22 2007-03-01 Critical Therapeutics, Inc. Method of quantitating nucleic acids by flow cytometry microparticle-based array
US20110281738A1 (en) 2005-10-07 2011-11-17 Callida Genomics, Inc. Self-assembled single molecule arrays and uses thereof
US7960104B2 (en) 2005-10-07 2011-06-14 Callida Genomics, Inc. Self-assembled single molecule arrays and uses thereof
WO2007081387A1 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices, methods of use, and kits for performing diagnostics
WO2007081385A2 (en) 2006-01-11 2007-07-19 Raindance Technologies, Inc. Microfluidic devices and methods of use in the formation and control of nanoreactors
US20100137163A1 (en) 2006-01-11 2010-06-03 Link Darren R Microfluidic Devices and Methods of Use in The Formation and Control of Nanoreactors
US20070172873A1 (en) 2006-01-23 2007-07-26 Sydney Brenner Molecular counting
WO2007089541A2 (en) 2006-01-27 2007-08-09 President And Fellows Of Harvard College Fluidic droplet coalescence
US20070195127A1 (en) 2006-01-27 2007-08-23 President And Fellows Of Harvard College Fluidic droplet coalescence
US20090264299A1 (en) 2006-02-24 2009-10-22 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20120135893A1 (en) 2006-02-24 2012-05-31 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20090118488A1 (en) 2006-02-24 2009-05-07 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20090005252A1 (en) 2006-02-24 2009-01-01 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20090155781A1 (en) 2006-02-24 2009-06-18 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US20070228588A1 (en) 2006-03-30 2007-10-04 Yasuko Noritomi Apparatus for producing particles, emulsifier holding member, method for producing particles, and method for producing molecular membrane
JP2007268350A (en) 2006-03-30 2007-10-18 Toshiba Corp Apparatus for producing fine particle, emulsifier holding part, method for producing fine particle and method for producing molecular film
WO2007114794A1 (en) 2006-03-31 2007-10-11 Nam Trung Nguyen Active control for droplet-based microfluidics
WO2007121489A2 (en) 2006-04-19 2007-10-25 Applera Corporation Reagents, methods, and libraries for gel-free bead-based sequencing
US20070264320A1 (en) 2006-05-09 2007-11-15 The Regents Of The University Of California Microfluidic device for forming monodisperse lipoplexes
US20080014589A1 (en) 2006-05-11 2008-01-17 Link Darren R Microfluidic devices and methods of use thereof
US20080003142A1 (en) 2006-05-11 2008-01-03 Link Darren R Microfluidic devices
US20130210639A1 (en) 2006-05-11 2013-08-15 Darren R. Link Microfluidic devices
WO2007133710A2 (en) 2006-05-11 2007-11-22 Raindance Technologies, Inc. Microfluidic devices and methods of use thereof
WO2007139766A2 (en) 2006-05-22 2007-12-06 Nanostring Technologies, Inc. Systems and methods for analyzing nanoreporters
WO2007140015A2 (en) 2006-05-26 2007-12-06 Althea Technologies, Inc Biochemical analysis of partitioned cells
WO2007138178A2 (en) 2006-05-30 2007-12-06 Centre National De La Recherche Scientifique Method for treating drops in a microfluid circuit
WO2007149432A2 (en) 2006-06-19 2007-12-27 The Johns Hopkins University Single-molecule pcr on microparticles in water-in-oil emulsions
US7666664B2 (en) 2006-07-14 2010-02-23 Roche Molecular Systems, Inc. Instrument for heating and cooling
WO2008021123A1 (en) 2006-08-07 2008-02-21 President And Fellows Of Harvard College Fluorocarbon emulsion stabilizing surfactants
US20100105112A1 (en) 2006-08-07 2010-04-29 Christian Holtze Fluorocarbon emulsion stabilizing surfactants
US20090035770A1 (en) 2006-10-25 2009-02-05 The Regents Of The University Of California Inline-injection microdevice and microfabricated integrated DNA analysis system using same
US7910354B2 (en) 2006-10-27 2011-03-22 Complete Genomics, Inc. Efficient arrays of amplified polynucleotides
US8603749B2 (en) 2006-11-15 2013-12-10 Biospherex, LLC Multitag sequencing ecogenomics analysis-US
US20140194323A1 (en) 2006-11-15 2014-07-10 BioSpherex, LLC a Limited Liability Company Multitag sequencing ecogenomics analysis-us
WO2008091792A2 (en) 2007-01-23 2008-07-31 Honeywell International Inc. Hydrogel microarray with embedded metal nanoparticles
US20080241820A1 (en) 2007-02-16 2008-10-02 Krutzik Peter O Multiplex cellular assays using detectable cell barcodes
US8003312B2 (en) 2007-02-16 2011-08-23 The Board Of Trustees Of The Leland Stanford Junior University Multiplex cellular assays using detectable cell barcodes
US20110263457A1 (en) 2007-02-16 2011-10-27 Krutzik Peter O Multiplex Cellular Assays Using Detectable Cell Barcodes
WO2008102057A1 (en) 2007-02-21 2008-08-28 Valtion Teknillinen Tutkimuskeskus Method and test kit for determining the amounts of target sequences and nucleotide variations therein
WO2008109176A2 (en) 2007-03-07 2008-09-12 President And Fellows Of Harvard College Assays and other reactions involving droplets
US9017948B2 (en) 2007-03-07 2015-04-28 President And Fellows Of Harvard College Assays and other reactions involving droplets
US20100136544A1 (en) 2007-03-07 2010-06-03 Jeremy Agresti Assays and other reactions involving droplets
US20140199731A1 (en) 2007-03-07 2014-07-17 President And Fellows Of Harvard College Assay and other reactions involving droplets
US20140199730A1 (en) 2007-03-07 2014-07-17 President And Fellows Of Harvard College Assays and other reactions involving droplets
US20090012187A1 (en) 2007-03-28 2009-01-08 President And Fellows Of Harvard College Emulsions and Techniques for Formation
EP2136786B1 (en) 2007-03-28 2012-10-10 The President and Fellows of Harvard College Apparatus for forming droplets
WO2008121342A2 (en) 2007-03-28 2008-10-09 President And Fellows Of Harvard College Emulsions and techniques for formation
US7776927B2 (en) 2007-03-28 2010-08-17 President And Fellows Of Harvard College Emulsions and techniques for formation
US20100130369A1 (en) 2007-04-23 2010-05-27 Advanced Liquid Logic, Inc. Bead-Based Multiplexed Analytical Methods and Instrumentation
WO2008134153A1 (en) 2007-04-23 2008-11-06 Advanced Liquid Logic, Inc. Bead-based multiplexed analytical methods and instrumentation
WO2009005680A1 (en) 2007-06-29 2009-01-08 President And Fellows Of Harvard College Methods and apparatus for manipulation of fluidic species
US20120015382A1 (en) 2007-07-13 2012-01-19 President And Fellows Of Harvard College Droplet-based selection
US20090068170A1 (en) 2007-07-13 2009-03-12 President And Fellows Of Harvard College Droplet-based selection
WO2009011808A1 (en) 2007-07-13 2009-01-22 President And Fellows Of Harvard College Droplet-based selection
US20090053169A1 (en) 2007-08-20 2009-02-26 Pharmain Corporation Oligonucleotide Core Carrier Compositions for Delivery of Nucleic Acid-Containing Therapeutic Agents, Methods of Making and Using the Same
US8268564B2 (en) 2007-09-26 2012-09-18 President And Fellows Of Harvard College Methods and applications for stitched DNA barcodes
US20090098555A1 (en) 2007-09-26 2009-04-16 President And Fellows Of Harvard College Methods and applications for stitched dna barcodes
WO2009061372A1 (en) 2007-11-02 2009-05-14 President And Fellows Of Harvard College Systems and methods for creating multi-phase entities, including particles and/or fluids
US20130157899A1 (en) 2007-12-05 2013-06-20 Perkinelmer Health Sciences, Inc. Reagents and methods relating to dna assays using amplicon probes on encoded particles
US20110033854A1 (en) 2007-12-05 2011-02-10 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
WO2009085215A1 (en) 2007-12-21 2009-07-09 President And Fellows Of Harvard College Systems and methods for nucleic acid sequencing
US20110267457A1 (en) 2007-12-21 2011-11-03 David A Weitz Systems and methods for nucleic acid sequencing
US20090202984A1 (en) 2008-01-17 2009-08-13 Sequenom, Inc. Single molecule nucleic acid sequence analysis processes and compositions
JP2009208074A (en) 2008-02-08 2009-09-17 Kao Corp Manufacturing method of fine particle dispersion liquid
US20100021984A1 (en) 2008-05-23 2010-01-28 Edd Jon F Microfluidic Droplet Encapsulation
US20140065234A1 (en) 2008-06-05 2014-03-06 President And Fellows Of Harvard College Polymersomes, liposomes, and other species associated with fluidic droplets
US20110305761A1 (en) 2008-06-05 2011-12-15 President And Fellows Of Harvard College Polymersomes, colloidosomes, liposomes, and other species associated with fluidic droplets
US20120172259A1 (en) 2008-07-02 2012-07-05 Illumina Cambridge Limited Using populations of beads for the fabrication of arrays on surfaces
WO2010004018A2 (en) 2008-07-11 2010-01-14 Eth Zurich Degradable microcapsules
US20100022414A1 (en) 2008-07-18 2010-01-28 Raindance Technologies, Inc. Droplet Libraries
US20100069263A1 (en) 2008-09-12 2010-03-18 Washington, University Of Sequence tag directed subassembly of short sequencing reads into long sequencing reads
WO2010033200A2 (en) 2008-09-19 2010-03-25 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US20110218123A1 (en) 2008-09-19 2011-09-08 President And Fellows Of Harvard College Creation of libraries of droplets and related species
US20110092376A1 (en) 2008-09-23 2011-04-21 Quantalife, Inc. System for droplet-based assays using an array of emulsions
US20100173394A1 (en) 2008-09-23 2010-07-08 Colston Jr Billy Wayne Droplet-based assay system
US20110086780A1 (en) 2008-09-23 2011-04-14 Quantalife, Inc. System for forming an array of emulsions
US20110092392A1 (en) 2008-09-23 2011-04-21 Quantalife, Inc. System for forming an array of emulsions
US20120015822A1 (en) 2008-12-19 2012-01-19 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
US8748094B2 (en) 2008-12-19 2014-06-10 President And Fellows Of Harvard College Particle-assisted nucleic acid sequencing
US20120121481A1 (en) 2009-03-13 2012-05-17 President And Fellows Of Harvard College Scale-up of flow-focusing microfluidic devices
WO2010148039A2 (en) 2009-06-15 2010-12-23 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
WO2010151776A2 (en) 2009-06-26 2010-12-29 President And Fellows Of Harvard College Fluid injection
US20120132288A1 (en) 2009-06-26 2012-05-31 President And Fellows Of Harvard College Fluid injection
US20120211084A1 (en) 2009-09-02 2012-08-23 President And Fellows Of Harvard College Multiple emulsions created using jetting and other techniques
WO2011047870A1 (en) 2009-10-22 2011-04-28 Plasticell Ltd Nested cell encapsulation
US20120222748A1 (en) 2009-10-27 2012-09-06 President And Fellows Of Harvard College Droplet creation techniques
WO2011056546A1 (en) 2009-10-27 2011-05-12 President And Fellows Of Harvard College Droplet creation techniques
US20120220497A1 (en) 2009-11-03 2012-08-30 Gen 9, Inc. Methods and Microfluidic Devices for the Manipulation of Droplets in High Fidelity Polynucleotide Assembly
GB2485850A (en) 2009-11-25 2012-05-30 Bio Rad Laboratories DNA copy number and Chromosome aneuploidy detection by amplification wherein the ligated products are partitioned into oil droplets prior to amplification
WO2011066476A1 (en) 2009-11-25 2011-06-03 Quantalife, Inc. Methods and compositions for detecting genetic material
US20110160078A1 (en) 2009-12-15 2011-06-30 Affymetrix, Inc. Digital Counting of Individual Molecules by Stochastic Attachment of Diverse Labels
US20130109575A1 (en) 2009-12-23 2013-05-02 Raindance Technologies, Inc. Microfluidic systems and methods for reducing the exchange of molecules between droplets
US20110217736A1 (en) 2010-03-02 2011-09-08 Quantalife, Inc. System for hot-start amplification via a multiple emulsion
US20120190032A1 (en) 2010-03-25 2012-07-26 Ness Kevin D Droplet generation for droplet-based assays
US20120000777A1 (en) 2010-06-04 2012-01-05 The Regents Of The University Of California Devices and methods for forming double emulsion droplet compositions and polymer particles
WO2012012037A1 (en) * 2010-07-19 2012-01-26 New England Biolabs, Inc. Oligonucleotide adaptors: compositions and methods of use
US20120071331A1 (en) 2010-09-21 2012-03-22 James Casbon Increasing confidence of allele calls with molecular counting
US20130028812A1 (en) 2010-10-07 2013-01-31 The Regents Of The University Of California Methods and systems for on demand droplet generation and impedance based detection
WO2012048341A1 (en) 2010-10-08 2012-04-12 President And Fellows Of Harvard College High-throughput single cell barcoding
US20130274117A1 (en) * 2010-10-08 2013-10-17 President And Fellows Of Harvard College High-Throughput Single Cell Barcoding
WO2012083225A2 (en) 2010-12-16 2012-06-21 Gigagen, Inc. System and methods for massively parallel analysis of nycleic acids in single cells
US20140057799A1 (en) 2010-12-16 2014-02-27 Gigagen System and Methods for Massively Parallel Analysis of Nucleic Acids in Single Cells
US20120196288A1 (en) * 2011-01-27 2012-08-02 Lawrence Livermore National Security, Llc Chip-Based Droplet Sorting
US20120220494A1 (en) 2011-02-18 2012-08-30 Raindance Technolgies, Inc. Compositions and methods for molecular labeling
US20150011430A1 (en) 2011-04-25 2015-01-08 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
WO2012149042A2 (en) 2011-04-25 2012-11-01 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US20120316074A1 (en) 2011-04-25 2012-12-13 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US20150011432A1 (en) 2011-04-25 2015-01-08 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US20140227706A1 (en) 2011-05-16 2014-08-14 Dna Chip Research Inc. Method for assessing progression of clinical state of malignant neoplasm by quantitative detection of DNA in blood
US20130046030A1 (en) 2011-05-23 2013-02-21 Basf Se Control of emulsions, including multiple emulsions
WO2012166425A2 (en) 2011-05-27 2012-12-06 President And Fellows Of Harvard College Methods of amplifying whole genome of a single cell
US20120309002A1 (en) 2011-06-02 2012-12-06 Raindance Technologies, Inc. Sample multiplexing
US20130189700A1 (en) 2011-07-25 2013-07-25 Bio-Rad Laboratories, Inc. Breakage of an emulsion containing nucleic acid
US20130079231A1 (en) 2011-09-09 2013-03-28 The Board Of Trustees Of The Leland Stanford Junior University Methods for obtaining a sequence
US20150111256A1 (en) 2012-02-17 2015-04-23 President And Fellows Of Harvard College Assembly of Nucleic Acid Sequences in Emulsions
US20130225418A1 (en) 2012-02-24 2013-08-29 Andrew Watson Labeling and sample preparation for sequencing
WO2013177220A1 (en) 2012-05-21 2013-11-28 The Scripps Research Institute Methods of sample preparation
US20140378349A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20150225777A1 (en) 2012-08-14 2015-08-13 10X Genomics, Inc. Capsule array devices and methods of use
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20140378350A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20150005199A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
US20150005200A1 (en) 2012-08-14 2015-01-01 10X Technologies, Inc. Compositions and methods for sample processing
WO2014028537A1 (en) 2012-08-14 2014-02-20 10X Technologies, Inc. Microcapsule compositions and methods
US20140378322A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
US20140155295A1 (en) 2012-08-14 2014-06-05 10X Technologies, Inc. Capsule array devices and methods of use
US20150224466A1 (en) 2012-08-14 2015-08-13 10X Genomics, Inc. Capsule array devices and methods of use
US20150225778A1 (en) 2012-08-14 2015-08-13 10X Genomics, Inc. Capsule array devices and methods of use
US20140206554A1 (en) 2012-12-14 2014-07-24 10X Technologies, Inc. Methods and Systems for Processing Polynucleotides
US20150218633A1 (en) 2012-12-14 2015-08-06 10X Genomics, Inc. Methods and Systems for Processing Polynucleotides
US20140227684A1 (en) 2013-02-08 2014-08-14 10X Technologies, Inc. Partitioning and processing of analytes and other species
US20140235506A1 (en) 2013-02-08 2014-08-21 10X Technologies, Inc. Polynucleotide barcode generation
US20150292988A1 (en) 2014-04-10 2015-10-15 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same

Non-Patent Citations (157)

* Cited by examiner, † Cited by third party
Title
Abate et al., Valve-based flow focusing for drog formation. Appl Phys Lett. 2009;94. 3 pages.
Abate, et al. High-throughput injection with microfluidics using picoinjectors. Proc Natl Acad Sci U S A. Nov. 9, 2010;107(45):19163-6. doi: 10.1073/pNas.1006888107. Epub Oct. 20, 2010.
Advisory Action dated Mar. 21, 2014 for U.S. Appl. No. 13/119,470.
Advisory Action dated May 16, 2014 for U.S. Appl. No. 13/503,588.
Advisory Action mailed Nov. 20, 2013 for U.S. Appl. No. 13/139,326.
Agresti, et al. Selection of ribozymes that catalyse multiple-turnover Diels-Alder cycloadditions by using in vitro compartmentalization. Proc Natl Acad Sci U S A. Nov. 8, 2005;102(45):16170-5. Epub Oct. 31, 2005.
Akselband, "Enrichment of slow-growing marine microorganisms from mixed cultures using gel microdrop (GMD) growth assay and fluorescence-activated cell sorting", J. Exp. Marine Bioi., 329: 196-205 (2006).
Akselband, "Rapid mycobacteria drug susceptibility testing using gel microdrop (GMD) growth assay and flow cytometry", J. Microbiol. Methods, 62:181-197 (2005).
Anna et al., "Formation of dispersions using 'flow focusing' in microchannels", Appln. Phys. Letts. 82:3 364 (2003).
Australian Office Action issued Dec. 17, 2013 for Application No. AU 2010315580.
Boone, et al. Plastic advances microfluidic devices. The devices debuted in silicon and glass, but plastic fabrication may make them hugely successful in biotechnology application. Analytical Chemistry. Feb. 2002; 78A-86A.
Braeckmans et al., Scanning the Code. Modern Drug Discovery. 2003:28-32.
Bransky, et al. A microfluidic droplet generator based on a piezoelectric actuator. Lab Chip. Feb. 21, 2009;9(4):516-20. doi: 10.1039/b814810d. Epub Nov. 20, 2008.
Carroll, "The selection of high-producing cell lines using flow cytometry and cell sorting", Exp. Op. Bioi. Therp., 4:11 1821-1829 (2004).
Chaudhary "A rapid method of cloning functioNal variable-region antibody genese in Escherichia coli as single-chain immunotoxins" Proc. Nat!. Acad. Sci USA 87: 1066-1070 (Feb. 1990).
Chechetkin et al., Sequencing by hybridization with the generic 6-mer oligonucleotide microarray: an advanced scheme for data processing. J Biomol Struct Dyn. Aug. 2000;18(1):83-101.
Chinese Office Action and search report mailed May 23, 2013 for Application No. CN 200880127116.4.
Chinese office action dated Jun. 18, 2012 for CN Application No. 200880127116.4.
Chou, et al. Disposable Microdevices for DNA Analysis and Cell Sorting. Proc. Solid-State Sensor and Actuator Workshop, Hilton Head, SC. Jun. 8-11, 1998; 11-14.
Clausell-Tormos et al., "Droplet-based microfluidic platforms for the encapsulation and screening of mammalian cells and multicellular organisms", Chem. Biol. 15:427-437 (2008).
De Bruin et al., UBS Investment Research. Q-Series®: DNa Sequencing. UBS Securities LLC. Jul. 12, 2007. 15 pages.
Demirci, et al. Single cell epitaxy by acoustic picolitre droplets. Lab Chip. Sep. 2007;7(9):1139-45. Epub Jul. 10, 2007.
Doerr, "The smallest bioreactor", Nature Methods, 2:5 326 (2005).
Dowding, et al. Oil core/polymer shell microcapsules by interNal phase separation from emulsion droplets. II: controlling the release profile of active molecules. Langmuir. Jun. 7, 2005;21(12):5278-84.
Drmanac et al., Sequencing by hybridization (SBH): advantages, achievements, and opportunities. Adv Biochem Eng Biotechnol. 2002;77 :75-101.
Droplet Based Sequencing (slides) dated (Mar. 12, 2008).
Esser-Kahn, et al. Triggered release from polymer capsules. Macromolecules. 2011; 44:5539-5553.
European office action dated Apr. 5, 2013 for Application No. EP 08865992.5.
European office action dated Aug. 29, 2013 for Application No. EP 08865992.5.
European office action dated Dec. 15, 2010 for EP Application No. 08865992.5.
European office action dated Jan. 23, 2012 for Application No. EP 08865992.5.
European search report and opinion dated Feb. 2, 2016 for EP Application No. 13829413.
Fisher, et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 2011;12(1):R1. doi: 10.1186/gb-2011-12-1-rl. Epub Jan. 4, 2011.
Freiberg, et al. Polymer microspheres for controlled drug release. Int J Pharm. Sep. 10, 2004;282(1-2):1-18.
Fu, "A micro fabricated fluorescence-activated cell sorter", Nature Biotech., 17:1109-1111 (1997).
Fulton et al., Advanced multiplexed analysis with the FlowMetrix system. Clin Chern. Sep. 1997;43(9): 1749-56.
Garstecki, et al. Formation of monodisperse bubbles in a microfluidic flow-focusing device. Applied Physics Letters. 2004; 85(13):2649-2651. DOI: 10.1063/1.1796526.
Gartner, et al. The Microfluidic Toolbox-examples for fluidic interfaces and standardization concepts. Proc. SPIE 4982, Microfluidics, BioMEMS, and Medical Microsystems, (Jan. 17, 2003); doi: 10.1117/12.479566.
Ghadessy, et al. Directed evolution of polymerase function by compartmentalized self-replication. Proc Natl Acad Sci U S A. Apr. 10, 2001;98(8):4552-7. Epub Mar. 27, 2001.
Gyarmati, et al. Reversible disulphide formation in polymer networks: a versatile functional group from synthesis to applications. European Polymer Journal. 2013;49:1268-1286.
Hashimshony, et al. CEL-Seq: Single-Cell RNa-Seq by Multiplexed Linear Amplification. Cell Rep. Sep. 27, 2012;2(3):666-73. doi: 10.1016/j.celrep.2012.08.003. Epub Aug. 30, 2012.
He "Selective Encapsulation of Single Cells and Subcellular Organelles into Picoliter- and Femtoliter-Volume Droplets" ANal. Chern 77: 1539-1544 (2005).
Holtze, et al. Biocompatible surfactants for water-in-fluorocarbon emulsions. Lab Chip. Oct. 2008;8(10):1632-9. doi: 10.1039/b806706f. Epub Sep. 2, 2008.
Huebner, "Quantitative detection of protein expression in single cells using droplet microfluidics", Chern. Commun 1218-1220 (2007).
Hug, et al. Measurement of the number of molecules of a single mRNA species in a complex mRNA preparation. J Theor Biol. Apr. 21, 2003;221(4):615-24.
International Preliminary Report on Patentability dated Jul. 1, 2010 for PCT/US2008/013912.
International Preliminary Report on Patentability dated Jun. 30, 2011 for PCT/US2009/006649.
International Preliminary Report on Patentability dated Mar. 31, 2011 for PCT/US09/005184.
International Preliminary Report on Patentability dated May 10, 2012 for PCT/US2010/054050.
International Preliminary Report on Patentability dated Sep. 17, 2009 for PCT/US2008/003185 mailed Sep. 17, 2009.
International search report and written opinion dated Apr. 3, 2009 for PCT/US2008/013912.
International search report and written opinion dated Aug. 16, 2010 for PCT/US2009/005184.
International search report and written opinion dated Aug. 19, 2015 for PCT/US2015/025197.
International search report and written opinion dated Aug. 20, 2014 for PCT/US2014/015424.
International search report and written opinion dated Dec. 16, 2013 for PCT/US2013/054797.
International search report and written opinion dated Jan. 12, 2009 for PCT/US2008/003185.
International search report and written opinion dated Jan. 31, 2011 for PCT/US2010/054050.
International search report and written opinion dated May 14, 2014 for PCT/US2014/015427.
International search report and written opinion dated May 14, 2015 for PCT/US2014/044398.
International search report and written opinion dated May 16, 2014 for PCT/US2013/074764.
International search report and written opinion dated Oct. 2, 2009 for PCT/US2009/004037.
International search report and written opinion dated Oct. 21, 2009 for PCT/US2009/003389.
International search report and written opinion dated Oct. 29, 2008 for PCT/US2008/008563.
International search report dated Apr. 22, 2009 for PCT/US2009/000664.
Japanese Final Rejection dated Aug. 5, 2014 for Application No. JP 2012-536941.
Japanese Office Action and mailed Jul. 17, 2013 for Application No. JP 2010-539498.
Japanese Office Action mailed Nov. 19, 2013 for Application No. JP 2012-536941.
Khomiakov A et al., [Analysis of perfect and mismatched DNA duplexes by a generic hexanucleotide microchip]. Mol Bioi (Mosk). Jul.-Aug. 2003;37(4):726-41. Russian. Abstract only.
Kim, et al. Albumin loaded microsphere of amphiphilic poly(ethylene glycol)/ poly(alpha-ester) multiblock copolymer. Eur J Pharm Sci. Nov. 2004;23(3):245-51.
Kim, et al. Fabrication of monodisperse gel shells and functioNal microgels in microfluidic devices. Angew Chem Int Ed Engl. 2007;46(11):1819-22.
Koster et al., "Drop-based microfluidic devices for encapsulation of single cells", Lab on a Chip The Royal Soc. of Chern. 8: 1110-1115 (2008).
Kutyavin, et al. Oligonucleotides containing 2-aminoadenine and 2-thiothymine act as selectively binding complementary agents. Biochemistry. Aug. 27, 1996;35(34):11170-6.
Li, Y., et al., "PEGylated PLGA Nanoparticles as protein carriers: synthesis, preparation and biodistribution in rats," JourNal of Controlled Release, vol. 71, pp. 203-211 (2001).
Liu, et al. Preparation of uniform-sized PLA microcapsules by combining Shirasu porous glass membrane emulsification technique and multiple emulsion-solvent evaporation method. J Control Release. Mar. 2, 2005;103(1):31-43. Epub Dec. 21, 2004.
Liu, et al. Smart thermo-triggered squirting capsules for Nanoparticle delivery. Soft Matter. 2010; 6(16):3759-3763.
Loscertales, I.G., et al., "Micro/Nano Encapsulation via Electrified Coaxial Liquid Jets," Science, vol. 295, pp. 1695-1698 (2002).
Love, "A microengraving method for rapid selection of single cells producing antigen-specific antibodies", Nature Biotech, 24:6 703 (Jun. 2006).
Marcus. Gene method offers diagnostic hope. The Wall Street JourNal. Jul. 11, 2012.
Mazutis, et al. Selective droplet coalescence using microfluidic systems. Lab Chip. Apr. 24, 2012;12(10):1800-6. doi: 10.1039/c21c40121e. Epub Mar. 27, 2012.
Merriman, et al. Progress in ion torrent semiconductor chip based sequencing. Electrophoresis. Dec. 2012;33(23):3397-417. doi: 10.1002/elps.201200424.
Microfluidic ChipShop. Microfluidic product catalogue. Mar. 2005.
Microfluidic ChipShop. Microfluidic product catalogue. Oct. 2009.
Mirzabekov, "DNA Sequencing by Hybridization-a Megasequencing Method and a Diagnostic Tool?" Trends in Biotechnology 12(1): 27-32 (1994).
Mouritzen et al., Single nucleotide polymorphism genotyping using locked nucleic acid (LNa). Expert Rev Mol Diagn. Jan. 2003;3(1):27-38.
Nguyen, et al. In situ hybridization to chromosomes stabilized in gel microdrops. Cytometry. 1995; 21:111-119.
Notice of Allowance dated Jan. 27, 2014 for U.S Appl. No. 13/139,326.
Oberholzer, et al. Polymerase chain reaction in liposomes. Chem Biol. Oct. 1995;2(10):677-82.
Office action dated Apr. 20, 2015 for U.S. Appl. No. 13/966,150.
Office action dated Apr. 24, 2013 for U.S. Appl. No. 13/119,470.
Office action dated Aug. 6, 2013 for U.S. Appl. No. 13/139,326.
Office action dated Aug. 6, 2014 for U.S. Appl. No. 12/529,926.
Office action dated Dec. 5, 2013 for U.S. Appl. No. 13/119,470.
Office action dated Feb. 10, 2014 for U.S. Appl. No. 13/503,588.
Office action dated Feb. 23, 2016 for U.S. Appl. No. 14/104,650.
Office action dated Feb. 28, 2013 for U.S. Appl. No. 13/139,326.
Office action dated Jan. 15, 2015 for U.S. Appl. No. 14/250,701.
Office action dated Jan. 29, 2016 for U.S. Appl. No. 14/175,935.
Office action dated Jan. 4, 2010 for U.S. Appl. No. 12/172,186.
Office action dated Jul. 30, 2014 for U.S. Appl. No. 12/809,120.
Office action dated Mar. 1, 2016 for U.S. Appl. No. 14/250,701.
Office action dated Mar. 14, 2016 for U.S. Appl. No. 14/680,808.
Office action dated May 20, 2014 for U.S. Appl. No. 14/172,266.
Office action dated May 20, 2014 for U.S. Appl. No. 14/172,326.
Office action dated May 28, 2013 for U.S. Appl. No. 12/529,926.
Office action dated Nov. 6, 2015 for U.S. Appl. No. 13/966,150.
Office action dated Oct. 1, 2012 for U.S. Appl. No. 12/529,926.
Office action dated Oct. 9, 2015 for U.S. Appl. No. 14/680,808.
Office action dated Sep. 10, 2014 for U.S. Appl. No. 14/250,701.
Office action dated Sep. 17, 2013 for U.S. Appl. No. 13/503,588.
Office action dated Sep. 25, 2015 for U.S. Appl. No. 14/250,701.
Office Action mailed Apr. 29, 2014 for EP Application No. 08865992.5.
Office Action mailed Dec. 16, 2013 for CN Application No. 201080055990.9.
Office Action mailed May 23, 2013 for CN Application No. 200880127116.4.
Ogawa, et al. Production and characterization of O/W emulsions containing cationic droplets stabilized by lecithin-chitosan membranes. J Agric Food Chem. Apr. 23, 2003;51(9):2806-12.
Okushima, "Controlled production of monodisperse double emulsions by two-step droplet breakup in microfluidic devices", Langmuir, 20:9905-9908 (2004).
Perez, C., et al., "Poly(lactic acid)-poly(ethylene glycol) Nanoparticles as new carriers for the delivery ofplasmid DNa," JourNal of Controlled Release, vol. 75, pp. 211-224 (2001).
Peters, et al. Accurate whole-genome sequencing and haplotyping from 10 to 20 human cells. Nature. Jul. 11, 2012;487(7406):190-5. doi: 10.1038/Nature11236.
Plunkett, et al. Chymotrypsin responsive hydrogel: application of a disulfide exchange protocol for the preparation of methacrylamide containing peptides. Biomacromolecules. Mar.-Apr. 2005;6(2):632-7.
Ryan, et al. Rapid assay for mycobacterial growth and antibiotic susceptibility using gel microdrop encapsulation. J Clin Microbiol. Jul. 1995;33(7):1720-6.
Schirinzi et al., Combinatorial sequencing-by-hybridization: aNalysis of the NFI gene. Genet Test. 2006 Spring;10(1):8-17.
Schmitt, "Bead-based multiplex genotyping of human papillomaviruses", J. Clinical Microbial., 44:2 504-512 (2006).
Shah, "Fabrication of mono disperse thermosensitive microgels and gel capsules in micro fluidic devices", Soft Matter, 4:2303-2309 (2008).
Shimkus, et al. A chemically cleavable biotinylated nucleotide: usefulness in the recovery of protein-DNA complexes from avidin affinity columns Proc Natl Acad Sci U S A. May 1985;82(9):2593-7.
Simeonov et al., Single nucleotide polymorphism genotyping using short, fluorescently labeled locked nucleic acid (LNa) probes and fluorescence polarization detection. Nucleic Acids Res. Sep. 1, 2002;30(17):e91.
Sorokin et al., DiscrimiNation between perfect and mismatched duplexes with oligonucleotide gel microchips: role of thermodyNamic and kinetic effects during hybridization. J Biomol Struct Dyn. Jun. 2005;22(6):725-34.
Su, et al., Microfluidics-Based Biochips: Technology Issues, Implementation Platforms, and Design-Automation Challenges. IEEE Transactions on Computer-Aided Design of Integrated Circuits and Systems. 2006;25(2):211-23. (Feb. 2006).
Sun et al., Progress in research and application of liquid-phase chip technology. Chinese JourNal Experimental Surgery. May 2005;22(5):639-40.
Tawfik, et al. Man-made cell-like compartments for molecular evolution. Nat Biotechnol. Jul. 1998;16(7):652-6.
Theberge, et al. Microdropelts in microfluidics: an evolving platform for discoveries in chemsitry and biology. Angew Chem Int Ed Engl. Aug. 9, 2010;49(34):5846-68. doi: 10.1002/anie.200906653.
Tubeleviciute, et al. Compartmentalized self-replication (CSR) selection of Thermococcus litoralis Sh1B DNa polymerase for diminished uracil binding. Protein Eng Des Sel. Aug. 2010;23(8):589-97. doi: 10.1093/protein/gzq032. Epub May 31, 2010.
U.S. Appl. No. 13/966,150, filed Aug. 13, 2013, Hindson et al.
U.S. Appl. No. 14/104,650, filed Dec. 12, 2013, Hindson et al.
U.S. Appl. No. 14/175,935, filed Feb. 7, 2014, Hindson et al.
U.S. Appl. No. 14/316,383, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/316,398, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/316,416, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/316,431, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/316,447, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/316,463, filed Jun. 26, 2014, Hindson et al.
U.S. Appl. No. 14/624,468, filed Feb. 17, 2015, Hindson et al.
U.S. Appl. No. 14/624,473, filed Feb. 17, 2015, Hindson et al.
U.S. Appl. No. 14/624,484, filed Feb. 17, 2015, Hindson et al.
U.S. Appl. No. 14/680,808, filed Apr. 7, 2015, Hindson et al.
U.S. Appl. No. 14/682,952, filed Apr. 8, 2015, Bharadwaj et al.
Wang et al., Single nucleotide polymorphism discrimiNation assisted by improved base stacking hybridization using oligonucleotide microarrays. Biotechniques. 2003;35:300-08.
Wang, et al. A novel thermo-induced self-bursting microcapsule with magnetic-targeting property. Chemphyschem. Oct. 5, 2009;10(14):2405-9.
Weaver, "Rapid clonal growth measurements at the single-cell level: gel microdroplets and flow cytometry", Biotechnology, 9:873-877 (1991).
Whitesides, "Soft lithography in biology and biochemistry", Annual Review of Biomedical Engineering, 3:335-373 (2001).
Williams, et al. Amplification of complex gene libraries by emulsion PCR. Nat Methods. Jul. 2006;3(7):545-50.
Woo, et al. G/C-modified oligodeoxynucleotides with selective complementarity: synthesis and hybridization properties. Nucleic Acids Res. Jul. 1, 1996;24(13):2470-5.
Xia, "Soft lithography", Annual Review of Material Science, 28: 153-184 (1998).
Yamamoto, et al. Chemical modification of Ce(IV)/EDTA-base artificial restriction DNa cutter for versatile manipulation of doulbe-stranded DNa. Nucleic Acids Research. 2007; 35(7):e53.
Zhang, "Combinatorial marking of cells and organelles with reconstituted fluorescent proteins", Cell, 119:137-144 (Oct. 1, 2004).
Zhang, et al. Degradable disulfide core-cross-linked micelles as a drug delivery system prepared from vinyl functioNalized nucleosides via the RAFT process. Biomacromolecules. Nov. 2008;9(11):3321-31. doi: 10.1021/bm800867n. Epub Oct. 9, 2008.
Zhao, J., et al., "Preparation of hemoglobin-loaded Nano-sized particles with porous structure as oxygen carriers," Biomaterials, vol. 28, pp. 1414-1422 (2007).
Zimmermann et at., Microscale production of hybridomas by hypo-osmolar electrofusion. Hum. Antibodies Hybridomas. Jan. 1992;3(1 ): 14-8.
Zong, et al. Genome-wide detection of single-nucleotide and copy-number variations of a single human cell. Science. Dec. 21, 2012;338(6114):1622-6. doi: 10.1126/science.1229164.

Cited By (170)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11591652B2 (en) 2010-12-16 2023-02-28 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
US10787706B2 (en) 2010-12-16 2020-09-29 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
US11053543B2 (en) 2010-12-16 2021-07-06 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
US10465243B2 (en) 2010-12-16 2019-11-05 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
US10106789B2 (en) 2010-12-16 2018-10-23 Gigagen, Inc. System and methods for massively parallel analysis of nucleic acids in single cells
US11001883B2 (en) 2012-03-05 2021-05-11 The General Hospital Corporation Systems and methods for epigenetic sequencing
US11047003B2 (en) 2012-03-05 2021-06-29 The General Hospital Corporation Systems and methods for epigenetic sequencing
US9689024B2 (en) 2012-08-14 2017-06-27 10X Genomics, Inc. Methods for droplet-based sample preparation
US10597718B2 (en) 2012-08-14 2020-03-24 10X Genomics, Inc. Methods and systems for sample processing polynucleotides
US10752950B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11441179B2 (en) 2012-08-14 2022-09-13 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10053723B2 (en) 2012-08-14 2018-08-21 10X Genomics, Inc. Capsule array devices and methods of use
US10450607B2 (en) 2012-08-14 2019-10-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11078522B2 (en) 2012-08-14 2021-08-03 10X Genomics, Inc. Capsule array devices and methods of use
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US11021749B2 (en) 2012-08-14 2021-06-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9695468B2 (en) 2012-08-14 2017-07-04 10X Genomics, Inc. Methods for droplet-based sample preparation
US10626458B2 (en) 2012-08-14 2020-04-21 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10221442B2 (en) 2012-08-14 2019-03-05 10X Genomics, Inc. Compositions and methods for sample processing
US11359239B2 (en) 2012-08-14 2022-06-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10584381B2 (en) 2012-08-14 2020-03-10 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11035002B2 (en) 2012-08-14 2021-06-15 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10273541B2 (en) 2012-08-14 2019-04-30 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10669583B2 (en) 2012-08-14 2020-06-02 10X Genomics, Inc. Method and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10612090B2 (en) 2012-12-14 2020-04-07 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10253364B2 (en) 2012-12-14 2019-04-09 10X Genomics, Inc. Method and systems for processing polynucleotides
US10227648B2 (en) 2012-12-14 2019-03-12 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11473138B2 (en) 2012-12-14 2022-10-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11421274B2 (en) 2012-12-14 2022-08-23 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9856530B2 (en) 2012-12-14 2018-01-02 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10676789B2 (en) 2012-12-14 2020-06-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10150964B2 (en) 2013-02-08 2018-12-11 10X Genomics, Inc. Partitioning and processing of analytes and other species
US11193121B2 (en) 2013-02-08 2021-12-07 10X Genomics, Inc. Partitioning and processing of analytes and other species
US9644204B2 (en) 2013-02-08 2017-05-09 10X Genomics, Inc. Partitioning and processing of analytes and other species
US10395758B2 (en) 2013-08-30 2019-08-27 10X Genomics, Inc. Sequencing methods
US9824068B2 (en) 2013-12-16 2017-11-21 10X Genomics, Inc. Methods and apparatus for sorting data
US10343166B2 (en) 2014-04-10 2019-07-09 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US9694361B2 (en) 2014-04-10 2017-07-04 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10071377B2 (en) 2014-04-10 2018-09-11 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10137449B2 (en) 2014-04-10 2018-11-27 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US10150117B2 (en) 2014-04-10 2018-12-11 10X Genomics, Inc. Fluidic devices, systems, and methods for encapsulating and partitioning reagents, and applications of same
US11052368B2 (en) 2014-04-21 2021-07-06 Vilnius University Systems and methods for barcoding nucleic acids
US10596541B2 (en) 2014-04-21 2020-03-24 President And Fellows Of Harvard College Systems and methods for barcoding nucleic acids
US10839939B2 (en) 2014-06-26 2020-11-17 10X Genomics, Inc. Processes and systems for nucleic acid sequence assembly
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10344329B2 (en) 2014-06-26 2019-07-09 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10337061B2 (en) 2014-06-26 2019-07-02 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10480028B2 (en) 2014-06-26 2019-11-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10457986B2 (en) 2014-06-26 2019-10-29 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10208343B2 (en) 2014-06-26 2019-02-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10760124B2 (en) 2014-06-26 2020-09-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11629344B2 (en) 2014-06-26 2023-04-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11133084B2 (en) 2014-06-26 2021-09-28 10X Genomics, Inc. Systems and methods for nucleic acid sequence assembly
US10041116B2 (en) 2014-06-26 2018-08-07 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10030267B2 (en) 2014-06-26 2018-07-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11713457B2 (en) 2014-06-26 2023-08-01 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11597964B2 (en) 2014-09-09 2023-03-07 The Broad Institute, Inc. Droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11566279B2 (en) 2014-09-09 2023-01-31 The Broad Institute, Inc. Droplet-based method and apparatus for composite single-cell nucleic acid analysis
US11739368B2 (en) 2014-10-29 2023-08-29 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US10287623B2 (en) 2014-10-29 2019-05-14 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequencing
US10245587B2 (en) 2014-11-05 2019-04-02 10X Genomics, Inc. Instrument systems for integrated sample processing
US9975122B2 (en) 2014-11-05 2018-05-22 10X Genomics, Inc. Instrument systems for integrated sample processing
US11135584B2 (en) 2014-11-05 2021-10-05 10X Genomics, Inc. Instrument systems for integrated sample processing
US10557158B2 (en) 2015-01-12 2020-02-11 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10221436B2 (en) 2015-01-12 2019-03-05 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US11414688B2 (en) 2015-01-12 2022-08-16 10X Genomics, Inc. Processes and systems for preparation of nucleic acid sequencing libraries and libraries prepared using same
US10650912B2 (en) 2015-01-13 2020-05-12 10X Genomics, Inc. Systems and methods for visualizing structural variation and phasing information
US10854315B2 (en) 2015-02-09 2020-12-01 10X Genomics, Inc. Systems and methods for determining structural variation and phasing using variant call data
US11603554B2 (en) 2015-02-24 2023-03-14 10X Genomics, Inc. Partition processing methods and systems
US11274343B2 (en) 2015-02-24 2022-03-15 10X Genomics, Inc. Methods and compositions for targeted nucleic acid sequence coverage
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US11873483B2 (en) 2015-03-11 2024-01-16 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
US11746367B2 (en) 2015-04-17 2023-09-05 President And Fellows Of Harvard College Barcoding systems and methods for gene sequencing and other applications
US11904310B2 (en) 2015-10-28 2024-02-20 The Broad Institute, Inc. High-throughput dynamic reagent delivery system
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US11371094B2 (en) 2015-11-19 2022-06-28 10X Genomics, Inc. Systems and methods for nucleic acid processing using degenerate nucleotides
US11873528B2 (en) 2015-12-04 2024-01-16 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11624085B2 (en) 2015-12-04 2023-04-11 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US10774370B2 (en) 2015-12-04 2020-09-15 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11473125B2 (en) 2015-12-04 2022-10-18 10X Genomics, Inc. Methods and compositions for nucleic acid analysis
US11081208B2 (en) 2016-02-11 2021-08-03 10X Genomics, Inc. Systems, methods, and media for de novo assembly of whole genome sequence data
US11084036B2 (en) 2016-05-13 2021-08-10 10X Genomics, Inc. Microfluidic systems and methods of use
US11667951B2 (en) 2016-10-24 2023-06-06 Geneinfosec, Inc. Concealing information present within nucleic acids
US10480029B2 (en) 2016-12-22 2019-11-19 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323278B2 (en) 2016-12-22 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10011872B1 (en) 2016-12-22 2018-07-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11248267B2 (en) 2016-12-22 2022-02-15 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11180805B2 (en) 2016-12-22 2021-11-23 10X Genomics, Inc Methods and systems for processing polynucleotides
US10954562B2 (en) 2016-12-22 2021-03-23 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10858702B2 (en) 2016-12-22 2020-12-08 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10793905B2 (en) 2016-12-22 2020-10-06 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11732302B2 (en) 2016-12-22 2023-08-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10550429B2 (en) 2016-12-22 2020-02-04 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11193122B2 (en) 2017-01-30 2021-12-07 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10428326B2 (en) 2017-01-30 2019-10-01 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
US11072816B2 (en) 2017-05-03 2021-07-27 The Broad Institute, Inc. Single-cell proteomic assay using aptamers
US11660601B2 (en) 2017-05-18 2023-05-30 10X Genomics, Inc. Methods for sorting particles
US10544413B2 (en) 2017-05-18 2020-01-28 10X Genomics, Inc. Methods and systems for sorting droplets and beads
US11898206B2 (en) 2017-05-19 2024-02-13 10X Genomics, Inc. Systems and methods for clonotype screening
US10927370B2 (en) 2017-05-26 2021-02-23 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11198866B2 (en) 2017-05-26 2021-12-14 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10844372B2 (en) 2017-05-26 2020-11-24 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11155810B2 (en) 2017-05-26 2021-10-26 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US10400235B2 (en) 2017-05-26 2019-09-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US11773389B2 (en) 2017-05-26 2023-10-03 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
US20220411859A1 (en) * 2017-08-01 2022-12-29 Illumina, Inc. Hydrogel beads for nucleotide sequencing
WO2019032760A1 (en) 2017-08-10 2019-02-14 Rootpath Genomics, Inc. Improved method to analyze nucleic acid contents from multiple biological particles
US10357771B2 (en) 2017-08-22 2019-07-23 10X Genomics, Inc. Method of producing emulsions
US10766032B2 (en) 2017-08-22 2020-09-08 10X Genomics, Inc. Devices having a plurality of droplet formation regions
US11565263B2 (en) 2017-08-22 2023-01-31 10X Genomics, Inc. Droplet forming devices and system with differential surface properties
US10821442B2 (en) 2017-08-22 2020-11-03 10X Genomics, Inc. Devices, systems, and kits for forming droplets
US10610865B2 (en) 2017-08-22 2020-04-07 10X Genomics, Inc. Droplet forming devices and system with differential surface properties
US10549279B2 (en) 2017-08-22 2020-02-04 10X Genomics, Inc. Devices having a plurality of droplet formation regions
US10583440B2 (en) 2017-08-22 2020-03-10 10X Genomics, Inc. Method of producing emulsions
US10898900B2 (en) 2017-08-22 2021-01-26 10X Genomics, Inc. Method of producing emulsions
US10590244B2 (en) 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US11441172B2 (en) 2017-10-04 2022-09-13 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US11884964B2 (en) 2017-10-04 2024-01-30 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10837047B2 (en) 2017-10-04 2020-11-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US11725231B2 (en) 2017-10-26 2023-08-15 10X Genomics, Inc. Methods and systems for nucleic acid preparation and chromatin analysis
US11833515B2 (en) 2017-10-26 2023-12-05 10X Genomics, Inc. Microfluidic channel networks for partitioning
US11584954B2 (en) 2017-10-27 2023-02-21 10X Genomics, Inc. Methods and systems for sample preparation and analysis
US11884962B2 (en) 2017-11-15 2024-01-30 10X Genomics, Inc. Functionalized gel beads
US10745742B2 (en) 2017-11-15 2020-08-18 10X Genomics, Inc. Functionalized gel beads
US10876147B2 (en) 2017-11-15 2020-12-29 10X Genomics, Inc. Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
US11365438B2 (en) 2017-11-30 2022-06-21 10X Genomics, Inc. Systems and methods for nucleic acid preparation and analysis
US10928386B2 (en) 2018-02-12 2021-02-23 10X Genomics, Inc. Methods and systems for characterizing multiple analytes from individual cells or cell populations
US10725027B2 (en) 2018-02-12 2020-07-28 10X Genomics, Inc. Methods and systems for analysis of chromatin
US11255847B2 (en) 2018-02-12 2022-02-22 10X Genomics, Inc. Methods and systems for analysis of cell lineage
US10816543B2 (en) 2018-02-12 2020-10-27 10X Genomics, Inc. Methods and systems for analysis of major histocompatability complex
US11739440B2 (en) 2018-02-12 2023-08-29 10X Genomics, Inc. Methods and systems for analysis of chromatin
US11131664B2 (en) 2018-02-12 2021-09-28 10X Genomics, Inc. Methods and systems for macromolecule labeling
US11002731B2 (en) 2018-02-12 2021-05-11 10X Genomics, Inc. Methods and systems for antigen screening
US11852628B2 (en) 2018-02-22 2023-12-26 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
US20210254051A1 (en) * 2018-04-03 2021-08-19 Guangzhou Burning Rock Dx Co., Ltd. Compositions and methods for preparing nucleic acid libraries
US11155881B2 (en) 2018-04-06 2021-10-26 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US11932899B2 (en) 2018-06-07 2024-03-19 10X Genomics, Inc. Methods and systems for characterizing nucleic acid molecules
US11703427B2 (en) 2018-06-25 2023-07-18 10X Genomics, Inc. Methods and systems for cell and bead processing
US11873530B1 (en) 2018-07-27 2024-01-16 10X Genomics, Inc. Systems and methods for metabolome analysis
US11459607B1 (en) 2018-12-10 2022-10-04 10X Genomics, Inc. Systems and methods for processing-nucleic acid molecules from a single cell using sequential co-partitioning and composite barcodes
US11358137B2 (en) 2018-12-26 2022-06-14 Industrial Technology Research Institute Tubular structure for producing droplets and method for producing droplets
US11845983B1 (en) 2019-01-09 2023-12-19 10X Genomics, Inc. Methods and systems for multiplexing of droplet based assays
US11851683B1 (en) 2019-02-12 2023-12-26 10X Genomics, Inc. Methods and systems for selective analysis of cellular samples
US11467153B2 (en) 2019-02-12 2022-10-11 10X Genomics, Inc. Methods for processing nucleic acid molecules
US11584953B2 (en) 2019-02-12 2023-02-21 10X Genomics, Inc. Methods for processing nucleic acid molecules
US11655499B1 (en) 2019-02-25 2023-05-23 10X Genomics, Inc. Detection of sequence elements in nucleic acid molecules
US11920183B2 (en) 2019-03-11 2024-03-05 10X Genomics, Inc. Systems and methods for processing optically tagged beads
WO2020207963A1 (en) 2019-04-12 2020-10-15 Miltenyi Biotec B.V. & Co. KG Conjugates having an enzymatically releasable detection moiety and a barcode moiety
US11351544B2 (en) 2019-10-10 2022-06-07 1859, Inc. Methods and systems for microfluidic screening
US11123735B2 (en) 2019-10-10 2021-09-21 1859, Inc. Methods and systems for microfluidic screening
US11919000B2 (en) 2019-10-10 2024-03-05 1859, Inc. Methods and systems for microfluidic screening
US11247209B2 (en) 2019-10-10 2022-02-15 1859, Inc. Methods and systems for microfluidic screening
US11351543B2 (en) 2019-10-10 2022-06-07 1859, Inc. Methods and systems for microfluidic screening
WO2021094421A1 (en) 2019-11-15 2021-05-20 Miltenyi Biotec B.V. & Co. KG Color and bardcoded beads for single cell indexing
US11512337B2 (en) 2020-01-13 2022-11-29 Fluent Biosciences Inc. Emulsion based drug screening
US11827936B2 (en) 2020-01-13 2023-11-28 Fluent Biosciences Inc. Methods and systems for single cell gene profiling
US11773452B2 (en) 2020-01-13 2023-10-03 Fluent Biosciences Inc. Single cell sequencing
US11866782B2 (en) 2020-03-16 2024-01-09 Fluent Biosciences Inc. Multi-omic analysis in monodisperse droplets
US11851700B1 (en) 2020-05-13 2023-12-26 10X Genomics, Inc. Methods, kits, and compositions for processing extracellular molecules
WO2022048780A1 (en) 2020-09-07 2022-03-10 Miltenyi Biotec B.V. & Co. KG Conjugates having an enzymatically releasable detection moiety and a barcode moiety
US11952626B2 (en) 2023-08-22 2024-04-09 10X Genomics, Inc. Probe-based analysis of nucleic acids and proteins

Also Published As

Publication number Publication date
CN105102697A (en) 2015-11-25
US10150964B2 (en) 2018-12-11
US11193121B2 (en) 2021-12-07
KR102190198B1 (en) 2020-12-14
EP2954104A1 (en) 2015-12-16
US20220154175A1 (en) 2022-05-19
AU2014214682B2 (en) 2018-07-26
US20140235506A1 (en) 2014-08-21
EP2954065A4 (en) 2016-08-24
JP2019107035A (en) 2019-07-04
US9644204B2 (en) 2017-05-09
EP3998342A1 (en) 2022-05-18
CA2900543C (en) 2023-01-31
CA2900543A1 (en) 2014-08-14
DK2954065T3 (en) 2021-09-06
EP2954104B1 (en) 2020-09-16
KR20200140929A (en) 2020-12-16
US10150963B2 (en) 2018-12-11
US20170342404A1 (en) 2017-11-30
KR20150119047A (en) 2015-10-23
WO2014124336A3 (en) 2014-10-23
US20140227684A1 (en) 2014-08-14
EP2954104A4 (en) 2016-08-24
CN108753766A (en) 2018-11-06
EP2954065B1 (en) 2021-07-28
CA2900481A1 (en) 2014-08-14
US20160304860A1 (en) 2016-10-20
JP2016511243A (en) 2016-04-14
JP2023063359A (en) 2023-05-09
BR112015019159A2 (en) 2017-07-18
WO2014124338A1 (en) 2014-08-14
KR20230003659A (en) 2023-01-06
JP2021038272A (en) 2021-03-11
US20140228255A1 (en) 2014-08-14
US20190276817A1 (en) 2019-09-12
WO2014124336A2 (en) 2014-08-14
EP2954065A2 (en) 2015-12-16
EP3862435A1 (en) 2021-08-11
AU2014214682A1 (en) 2015-08-13
US20170362587A1 (en) 2017-12-21

Similar Documents

Publication Publication Date Title
US11473138B2 (en) Methods and systems for processing polynucleotides
US9388465B2 (en) Polynucleotide barcode generation
US11421274B2 (en) Methods and systems for processing polynucleotides
US11591637B2 (en) Compositions and methods for sample processing
EP2931919B1 (en) Methods and systems for processing polynucleotides
EP3467160A1 (en) Compositions and methods for sample processing
CN113166807A (en) Nucleotide sequence generation by barcode bead co-localization in partitions
US20240002929A1 (en) Methods and systems for processing polynucleotides

Legal Events

Date Code Title Description
AS Assignment

Owner name: 10X TECHNOLOGIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HINDSON, BENJAMIN;JAROSZ, MIRNA;HARDENBOL, PAUL;AND OTHERS;SIGNING DATES FROM 20140214 TO 20140225;REEL/FRAME:033025/0665

AS Assignment

Owner name: 10X GENOMICS, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:10X TECHNOLOGIES, INC.;REEL/FRAME:034795/0154

Effective date: 20141106

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8